1
|
Smith FM, Kosman DJ. Brain microvascular endothelial cells differentiated from a Friedreich's Ataxia patient iPSC are deficient in tight junction protein expression and paracellularly permeable. Front Mol Neurosci 2025; 18:1511388. [PMID: 40303283 PMCID: PMC12037585 DOI: 10.3389/fnmol.2025.1511388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/19/2025] [Indexed: 05/02/2025] Open
Abstract
Friedreich's Ataxia (FA) is a rare, inherited ataxia resulting from GAA triplet expansions in the first intron of the Frataxin (FXN) gene, which encodes a mitochondrial protein involved in the incorporation of iron into iron-sulfur clusters. We previously identified decreased levels of F-actin and tight junction (TJ) proteins, which coincided with paracellular permeability in an FXN shRNA-mediated knockdown immortalized human brain microvascular endothelial cell (BMVEC) model. This premise is underexplored in the FA literature, prompting us to confirm these findings using a patient-derived iPSC model. One line each of FA patient iPSCs and age- and sex-matched apparently healthy iPSCs were differentiated into BMVEC-like cells. We quantified actin glutathionylation, F-actin abundance, TJ expression and organization, and barrier integrity. In the absence of dysregulated F-actin organization, FA iBMVEC exhibited a loss of 50% ZO-1, 63% Occludin, and 19% Claudin-5 protein expression, along with a disruption in the bi-cellular organization of the latter two proteins. Functionally, this correlated with barrier hyperpermeability, delayed barrier maturation, and increased flux of the fluorescent tracer Lucifer Yellow. These data indicate that decreased barrier integrity is a pathophysiological phenotype of FA brain microvascular endothelial cells. Clinically, this may represent a targetable pathway to reduce brain iron accumulation, neuroinflammation, and neurodegeneration profiles in FA. Additionally, an investigation into other barrier systems, such as the blood-nerve barrier, blood-CSF barrier, or cardiac vasculature, may provide insights into the extra-neural symptoms experienced by FA patients.
Collapse
Affiliation(s)
| | - Daniel J. Kosman
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
2
|
Bernardo VS, Torres FF, Zucão ACA, Chaves NA, Santana ILR, da Silva DGH. Disrupted homeostasis in sickle cells: Expanding the comprehension of metabolism adaptation and related therapeutic strategies. Tissue Cell 2025; 93:102717. [PMID: 39805212 DOI: 10.1016/j.tice.2024.102717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/02/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025]
Abstract
Sickle cell disease (SCD) is a hereditary hemolytic anemia associated with the alteration of the membrane composition of the sickle erythrocytes, the loss of glycolysis, dysregulation of the pyruvate phosphatase pathway, and changes in nucleotide metabolism of the sickle red blood cell (RBC). This review provides a comprehensive overview of the impact of the presence of Hb S, which leads to the disruption of the normal RBC metabolism. The intricate interplay between the redox and energetic balance in erythrocytic cells, where the glycolysis, pentose phosphate pathway, and methemoglobin reductase pathways are all altered in sickle RBC, is a key focus. Moreover, this review summarizes the current knowledge about the disease-modifying agents and their action mechanisms based on the sickle RBC alterations previously mentioned (i.e., their association with beneficial effects on the sickle cells' membrane, to their RBCs' energy metabolism, and to their oxidative status). Therefore, providing a comprehensive understanding of how sickle cells cope with the disruption of metabolic homeostasis and the most promising therapeutic agents able to ameliorate the various consequences of abnormal sickle RBC alterations.
Collapse
Affiliation(s)
| | | | | | - Nayara Alves Chaves
- Department of Biology, Universidade Estadual Paulista (UNESP), São Paulo, Brazil
| | | | - Danilo Grünig Humberto da Silva
- Department of Biology, Universidade Estadual Paulista (UNESP), São Paulo, Brazil; Campus de Três Lagoas, Universidade Federal de Mato Grosso do Sul (CPTL/UFMS), Mato Grosso do Sul, Brazil.
| |
Collapse
|
3
|
Lee L, Okudaira N, Murase K, Kong R, Jones HM. Determination of Vatiquinone Drug-Drug Interactions, as CYP450 Perpetrator and Victim, Using Physiologically Based Pharmacokinetic (PBPK) Modeling and Simulation. J Clin Pharmacol 2025; 65:160-169. [PMID: 39308341 PMCID: PMC11771645 DOI: 10.1002/jcph.6133] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 08/26/2024] [Indexed: 01/30/2025]
Abstract
Vatiquinone, a 15-lipoxygenase inhibitor, is in development for patients with Friedreich's ataxia. Physiologically based pharmacokinetic (PBPK) modeling addressed drug-drug interaction gaps without additional studies. A PBPK model (Simcyp Simulator version 21, full model) was developed using parameters obtained from in vitro studies, in silico estimation and optimization, and two clinical studies. A venous blood dosing model best characterized vatiquinone lymphatic absorption. Apparent oral clearance (CL/F) was used to optimize intrinsic clearance (CLint). Intestinal availability (Fg) was estimated using the hybrid flow term (Qgut), unbound fraction in the enterocytes (fugut), and gut intrinsic metabolic clearance (CLuG,int). Renal clearance (CLR) was set to zero. Assuming an Fa of 1, CYP3A4 contribution (fmCYP3A4) was further optimized. The PBPK model was verified with two clinical studies and demonstrated that it adequately characterized vatiquinone PK. As a perpetrator, the model predicted no risk for vatiquinone to significantly alter the drug exposures of CYP3A4 and CYP1A2 substrates as evident bynegligible reduction in both midazolam and caffeine area under the curve (AUC)inf and Cmax. As a victim, the model predicted that vatiquinone exposures are weakly influenced by moderate CYP3A4 inhibitors and inducers. With fluconazole coadministration, vatiquinone AUCinf and Cmax increased by nearly 50% and 25%, respectively. With efavirenz coadministration, vatiquinone AUCinf and Cmax decreased by approximately 20% and 10%, respectively. Results suggested that vatiquinone does not significantly impact CYP3A4 and CYP1A2 substrates and that moderate CYP3A4 inhibitors and inducers weakly impact vatiquinone AUC.
Collapse
Affiliation(s)
- Lucy Lee
- PTC Therapeutics, Inc.WarrenNJUSA
| | | | | | | | | |
Collapse
|
4
|
Lee L, Flach S, Xue H, Arivelu L, Golden L, Kong R, Darpo B. Lack of Concentration-QTc Relationship and Cardiac Risk With Vatiquinone Therapeutic and Supratherapeutic Doses. Clin Pharmacol Drug Dev 2024; 13:1227-1238. [PMID: 39415654 DOI: 10.1002/cpdd.1476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Vatiquinone, a 15-lipoxygenase inhibitor, is in development for patients with Friedreich ataxia. The study determined the effect of vatiquinone on electrocardiogram parameters. This was a 2-part, single-center, randomized, double-blinded, and placebo-controlled study. Part 1 used an adaptive approach to identify a supratherapeutic dose, while Part 2 evaluated the effect of vatiquinone on Fridericia corrected QT interval (QTcF). A safe and tolerated supratherapeutic dose of 1400 mg was identified. Concentration-QTcF analysis confirmed there was no statistically significant relationship between vatiquinone concentration and QTcF. QTcF effect (ie, ΔΔQTcF) exceeding 10 milliseconds was excluded for concentrations up to approximately 11,500 ng/mL. By-time-point analysis confirmed that least-squares mean ΔΔQTcF was below 10 milliseconds. Largest least-squares mean ΔΔQTcF of 1.5 milliseconds was observed at 2 hours after dosing. Vatiquinone did not have a clinically relevant effect on heart rate nor on cardiac conduction (PR interval and QRS interval). No new safety signals were found, as safety data are consistent with the known safety profile of vatiquinone. These findings altogether demonstrated that there is a minimal cardiac risk for vatiquinone concentrations up to the supratherapeutic dose level.
Collapse
Affiliation(s)
- Lucy Lee
- PTC Therapeutics, Inc., Warren, NJ, USA
| | | | | | | | | | | | | |
Collapse
|
5
|
Lee L, Thoolen M, Ma J, Kaushik D, Golden L, Kong R. Effect of Itraconazole, a CYP3A4 Inhibitor, and Rifampin, a CYP3A4 Inducer, on the Pharmacokinetics of Vatiquinone. Clin Pharmacol Drug Dev 2024. [PMID: 39133029 DOI: 10.1002/cpdd.1461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024]
Abstract
Vatiquinone is a small molecule inhibitor of 15-lipoxygenase in development for patients with Friedreich's ataxia. The objective of this analysis was to determine the effect of a cytochrome P450 isoform 3A4 (CYP3A4) inhibitor and inducer on vatiquinone pharmacokinetics (PKs). The coadministration of 400 mg of vatiquinone with 200 mg of itraconazole (a CYP3A4 inhibitor) resulted in increased maximum observed concentration (Cmax) of vatiquinone and systemic exposure (AUC0-inf) by approximately 3.5- and 2.9-fold, respectively. The coadministration of 400 mg of vatiquinone with 600 mg of rifampin (a CYP3A4 inducer) resulted in decreased vatiquinone Cmax and AUC0-inf by approximately 0.64- and 0.54-fold, respectively. The terminal half-life of vatiquinone was not affected by itraconazole or rifampin. These clinical study results confirm the in vitro reaction phenotyping data that shows that CYP3A4 plays an important role in vatiquinone metabolism. The result of this analysis together with phase 3 efficacy and safety data, population PK analysis, and the exposure-response relationship will determine if the extent of vatiquinone changes in the presence of CYP3A4 inhibitors and inducers are considered clinically relevant.
Collapse
Affiliation(s)
- Lucy Lee
- PTC Therapeutics, Inc., Warren, NJ, USA
| | | | - Jiyuan Ma
- PTC Therapeutics, Inc., Warren, NJ, USA
| | | | | | | |
Collapse
|
6
|
Chang KH, Chen CM. The Role of NRF2 in Trinucleotide Repeat Expansion Disorders. Antioxidants (Basel) 2024; 13:649. [PMID: 38929088 PMCID: PMC11200942 DOI: 10.3390/antiox13060649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Trinucleotide repeat expansion disorders, a diverse group of neurodegenerative diseases, are caused by abnormal expansions within specific genes. These expansions trigger a cascade of cellular damage, including protein aggregation and abnormal RNA binding. A key contributor to this damage is oxidative stress, an imbalance of reactive oxygen species that harms cellular components. This review explores the interplay between oxidative stress and the NRF2 pathway in these disorders. NRF2 acts as the master regulator of the cellular antioxidant response, orchestrating the expression of enzymes that combat oxidative stress. Trinucleotide repeat expansion disorders often exhibit impaired NRF2 signaling, resulting in inadequate responses to excessive ROS production. NRF2 activation has been shown to upregulate antioxidative gene expression, effectively alleviating oxidative stress damage. NRF2 activators, such as omaveloxolone, vatiquinone, curcumin, sulforaphane, dimethyl fumarate, and resveratrol, demonstrate neuroprotective effects by reducing oxidative stress in experimental cell and animal models of these diseases. However, translating these findings into successful clinical applications requires further research. In this article, we review the literature supporting the role of NRF2 in the pathogenesis of these diseases and the potential therapeutics of NRF2 activators.
Collapse
Affiliation(s)
- Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Kueishan, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Kueishan, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
7
|
Smith FM, Kosman DJ. Loss of filamentous actin, tight junction protein expression, and paracellular barrier integrity in frataxin-deficient human brain microvascular endothelial cells-implications for blood-brain barrier physiology in Friedreich's ataxia. Front Mol Biosci 2024; 10:1299201. [PMID: 38274097 PMCID: PMC10808331 DOI: 10.3389/fmolb.2023.1299201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction: Friedreich's Ataxia (FRDA) is the most prevalent inherited ataxia. FRDA results from loss of Frataxin (FXN), an essential mitochondrial iron trafficking protein. FRDA starts with an early burst of neurodegeneration of the dorsal root ganglion and cerebellar dentate nuclei, followed by progressive brain iron accumulation in the latter. End stage disease includes cardiac fibrosis that contributes to hypertrophic cardiomyopathy. The microvasculature plays an essential barrier role in both brain and heart homeostasis, thus an investigation of this tissue system in FRDA is essential to the delineation of the cellular dysfunction in this genetic disorder. Previous reports have identified cytoskeletal alterations in non-barrier forming FRDA cell models, but physiological consequences are limited. Methods: We investigated brain microvascular endothelial cell integrity in FRDA in a model of the blood-brain barrier (BBB). We have knocked down FXN in immortalized human brain microvascular endothelial cells (hBMVEC), which compose the microcapillaries of the BBB, by using shRNA. We confirmed known cellular pathophysiologies of FXN-knockdown including decreased energy metabolism, markers of oxidative stress, and increased cell size. Results: We investigated cytoskeletal architecture, identifying decreased filamentous actin and Occludin and Claudin-5 tight junction protein expression in shFXN hBMVECs. This was consistent with decreased transendothelial electrical resistance (TEER) and increased paracellular tracer flux during early barrier formation. shFXN hBMVEC start with only 67% barrier integrity of the controls, and flux a paracellular tracer at 800% of physiological levels. Discussion: We identified that insufficient FXN levels in the hBMVEC BBB model causes changes in cytoskeletal architecture and tight junction protein abundance, co-incident with increased barrier permeability. Changes in the integrity of the BBB may be related to patient brain iron accumulation, neuroinflammation, neurodegeneration, and stroke. Furthermore, our findings implicate other barrier cells, e.g., the cardiac microvasculature, loci of disease pathology in FRDA.
Collapse
Affiliation(s)
- Frances M. Smith
- Jacobs School of Medicine and Biomedical Sciences, Department of Biochemistry, The State University of New York at Buffalo, Buffalo, NY, United States
| | | |
Collapse
|
8
|
Scirè A, Casari G, Romaldi B, de Bari L, Antognelli C, Armeni T. Glutathionyl Hemoglobin and Its Emerging Role as a Clinical Biomarker of Chronic Oxidative Stress. Antioxidants (Basel) 2023; 12:1976. [PMID: 38001829 PMCID: PMC10669486 DOI: 10.3390/antiox12111976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Hemoglobin is one of the proteins that are more susceptible to S-glutathionylation and the levels of its modified form, glutathionyl hemoglobin (HbSSG), increase in several human pathological conditions. The scope of the present review is to provide knowledge about how hemoglobin is subjected to S-glutathionylation and how this modification affects its functionality. The different diseases that showed increased levels of HbSSG and the methods used for its quantification in clinical investigations will be also outlined. Since there is a growing need for precise and reliable methods for markers of oxidative stress in human blood, this review highlights how HbSSG is emerging more and more as a good indicator of severe oxidative stress but also as a key pathogenic factor in several diseases.
Collapse
Affiliation(s)
- Andrea Scirè
- Department of Life and Environmental Sciences (Di.S.V.A.), Università Politecnica delle Marche, 60131 Ancona, Italy
| | - Giulia Casari
- Department of Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy; (G.C.); (B.R.); (T.A.)
| | - Brenda Romaldi
- Department of Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy; (G.C.); (B.R.); (T.A.)
| | - Lidia de Bari
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), 70126 Bari, Italy;
| | - Cinzia Antognelli
- Department of Medicine and Surgery, Università Degli Studi di Perugia, 06129 Perugia, Italy;
| | - Tatiana Armeni
- Department of Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy; (G.C.); (B.R.); (T.A.)
| |
Collapse
|
9
|
Costa I, Barbosa DJ, Silva V, Benfeito S, Borges F, Remião F, Silva R. Research Models to Study Ferroptosis's Impact in Neurodegenerative Diseases. Pharmaceutics 2023; 15:pharmaceutics15051369. [PMID: 37242612 DOI: 10.3390/pharmaceutics15051369] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Ferroptosis is a type of regulated cell death promoted by the appearance of oxidative perturbations in the intracellular microenvironment constitutively controlled by glutathione peroxidase 4 (GPX4). It is characterized by increased production of reactive oxygen species, intracellular iron accumulation, lipid peroxidation, inhibition of system Xc-, glutathione depletion, and decreased GPX4 activity. Several pieces of evidence support the involvement of ferroptosis in distinct neurodegenerative diseases. In vitro and in vivo models allow a reliable transition to clinical studies. Several in vitro models, including differentiated SH-SY5Y and PC12 cells, among others, have been used to investigate the pathophysiological mechanisms of distinct neurodegenerative diseases, including ferroptosis. In addition, they can be useful in the development of potential ferroptosis inhibitors that can be used as disease-modifying drugs for the treatment of such diseases. On the other hand, in vivo models based on the manipulation of rodents and invertebrate animals, such as Drosophila melanogaster, Caenorhabditis elegans, and zebrafish, have been increasingly used for research in neurodegeneration. This work provides an up-to-date review of the main in vitro and in vivo models that can be used to evaluate ferroptosis in the most prevalent neurodegenerative diseases, and to explore potential new drug targets and novel drug candidates for effective disease-modifying therapies.
Collapse
Affiliation(s)
- Inês Costa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Daniel José Barbosa
- TOXRUN-Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Vera Silva
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Sofia Benfeito
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Fernanda Borges
- CIQUP-IMS-Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Renata Silva
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
10
|
Costa I, Barbosa DJ, Benfeito S, Silva V, Chavarria D, Borges F, Remião F, Silva R. Molecular mechanisms of ferroptosis and their involvement in brain diseases. Pharmacol Ther 2023; 244:108373. [PMID: 36894028 DOI: 10.1016/j.pharmthera.2023.108373] [Citation(s) in RCA: 156] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
Ferroptosis is a type of regulated cell death characterized by intracellular accumulation of iron and reactive oxygen species, inhibition of system Xc-, glutathione depletion, nicotinamide adenine dinucleotide phosphate oxidation and lipid peroxidation. Since its discovery and characterization in 2012, many efforts have been made to reveal the underlying mechanisms, modulating compounds, and its involvement in disease pathways. Ferroptosis inducers include erastin, sorafenib, sulfasalazine and glutamate, which, by inhibiting system Xc-, prevent the import of cysteine into the cells. RSL3, statins, Ml162 and Ml210 induce ferroptosis by inhibiting glutathione peroxidase 4 (GPX4), which is responsible for preventing the formation of lipid peroxides, and FIN56 and withaferin trigger GPX4 degradation. On the other side, ferroptosis inhibitors include ferrostatin-1, liproxstatin-1, α-tocopherol, zileuton, FSP1, CoQ10 and BH4, which interrupt the lipid peroxidation cascade. Additionally, deferoxamine, deferiprone and N-acetylcysteine, by targeting other cellular pathways, have also been classified as ferroptosis inhibitors. Increased evidence has established the involvement of ferroptosis in distinct brain diseases, including Alzheimer's, Parkinson's and Huntington's diseases, amyotrophic lateral sclerosis, multiple sclerosis, and Friedreich's ataxia. Thus, a deep understanding of how ferroptosis contributes to these diseases, and how it can be modulated, can open a new window of opportunities for novel therapeutic strategies and targets. Other studies have shown a sensitivity of cancer cells with mutated RAS to ferroptosis induction and that chemotherapeutic agents and ferroptosis inducers synergize in tumor treatment. Thus, it is tempting to consider that ferroptosis may arise as a target mechanistic pathway for the treatment of brain tumors. Therefore, this work provides an up-to-date review on the molecular and cellular mechanisms of ferroptosis and their involvement in brain diseases. In addition, information on the main ferroptosis inducers and inhibitors and their molecular targets is also provided.
Collapse
Affiliation(s)
- Inês Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Daniel José Barbosa
- TOXRUN - Toxicology Research Unit, Department of Sciences, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Sofia Benfeito
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal.
| | - Vera Silva
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Daniel Chavarria
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Fernanda Borges
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Renata Silva
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
11
|
Lee L, Murase K, Ma J, Thoolen M. Clinical Drug-Drug Interaction Between Vatiquinone, a 15-Lipoxygenase Inhibitor, and Rosuvastatin, a Breast Cancer Resistance Protein Substrate. Clin Pharmacol Drug Dev 2023; 12:279-286. [PMID: 36478438 DOI: 10.1002/cpdd.1199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/11/2022] [Indexed: 12/12/2022]
Abstract
Vatiquinone is a small-molecule inhibitor of 15-lipoxygenase in phase 3 development for patients with mitochondrial disease and Friedreich ataxia. The objective of this analysis was to determine the effect of vatiquinone on the pharmacokinetic profile of rosuvastatin, a breast cancer resistance protein substrate. In vitro investigations demonstrated potential inhibition of BCRP by vatiquinone (half maximal inhibitory concentration, 3.8 µM). An open-label, fixed-sequence drug-drug interaction study in healthy volunteers was conducted to determine the clinical relevance of this finding. Subjects received a single dose of 20-mg rosuvastatin followed by a 7-day washout. On days 8 through 14, subjects received 400 mg of vatiquinone 3 times daily. On day 12, subjects concomitantly received a single dose of 20-mg rosuvastatin. The geometric mean ratio for maximum plasma concentration was 77.8%; however, the rosuvastatin disposition phase appeared unaffected. The geometric mean ratios for the area under the plasma concentration-time curve from time 0 to time t and from time 0 to infinity were 103.2% and 99.9%, respectively. Mean rosuvastatin apparent elimination half-life was similar between treatment groups. These results demonstrate that vatiquinone has no clinically relevant effect on the pharmacokinetics of rosuvastatin.
Collapse
Affiliation(s)
- Lucy Lee
- PTC Therapeutics, Inc., South Plainfield, New Jersey, USA
| | | | - Jiyuan Ma
- PTC Therapeutics, Inc., South Plainfield, New Jersey, USA
| | - Martin Thoolen
- PTC Therapeutics, Inc., South Plainfield, New Jersey, USA
| |
Collapse
|
12
|
Smith FM, Kosman DJ. Frataxin-deficient human brain microvascular endothelial cells lose polymerized actin and are paracellularly permeable -implications for blood-brain barrier integrity in Friedreich's Ataxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.09.527936. [PMID: 36798283 PMCID: PMC9934603 DOI: 10.1101/2023.02.09.527936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Background Friedreich's Ataxia (FRDA) is the most prevalent inherited ataxia; the disease results from loss of Frataxin, an essential mitochondrial iron trafficking protein. FRDA presents as neurodegeneration of the dorsal root ganglion and cerebellar dentate nuclei, followed by brain iron accumulation in the latter. End stage disease includes cardiac fibrosis that contributes to hypertrophic cardiomyopathy. The microvasculature plays an essential barrier role in both the brain and heart, thus an investigation of this tissue system in FRDA is essential to the delineation of the cellular dysfunction in this genetic disorder. Here, we investigate brain microvascular endothelial cell integrity in FRDA in a model of the blood-brain barrier (BBB). Methods We used lentiviral mediated shRNA delivery to generate a novel FRDA model in immortalized human brain microvascular endothelial cells (hBMVEC) that compose the microcapillaries of the BBB. We verified known cellular pathophysiologies of FXN knockdown including increased oxidative stress, loss of energy metabolism, and increased cell size. Furthermore, we investigated cytoskeletal architecture including the abundance and organization of filamentous actin, and barrier physiology via transendothelial electrical resistance and fluorescent tracer flux. Results shFXN hBMVEC display the known FRDA cell morbidity including increased oxidative stress, decreased energy metabolism, and an increase in cell size. We demonstrate that shFXN hBMVEC have less overall filamentous actin, and that filamentous actin is lost at the cell membrane and cortical actin ring. Consistent with loss of cytoskeletal structure and anchorage, we found decreased barrier strength and increased paracellular tracer flux in the shFXN hBMVEC transwell model. Conclusion We identified that insufficient FXN levels in the hBMVEC BBB model causes changes in cytoskeletal architecture and increased barrier permeability, cell pathologies that may be related to patient brain iron accumulation, neuroinflammation, neurodegeneration, and stroke. Our findings implicate other barrier cells, e.g., the cardiac microvasculature, likely contributory also to disease pathology in FRDA.
Collapse
Affiliation(s)
- Frances M Smith
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, The University of New York at Buffalo
| | - Daniel J Kosman
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, The University of New York at Buffalo
| |
Collapse
|
13
|
Vrettou S, Wirth B. S-Glutathionylation and S-Nitrosylation in Mitochondria: Focus on Homeostasis and Neurodegenerative Diseases. Int J Mol Sci 2022; 23:15849. [PMID: 36555492 PMCID: PMC9779533 DOI: 10.3390/ijms232415849] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/24/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Redox post-translational modifications are derived from fluctuations in the redox potential and modulate protein function, localization, activity and structure. Amongst the oxidative reversible modifications, the S-glutathionylation of proteins was the first to be characterized as a post-translational modification, which primarily protects proteins from irreversible oxidation. However, a growing body of evidence suggests that S-glutathionylation plays a key role in core cell processes, particularly in mitochondria, which are the main source of reactive oxygen species. S-nitrosylation, another post-translational modification, was identified >150 years ago, but it was re-introduced as a prototype cell-signaling mechanism only recently, one that tightly regulates core processes within the cell’s sub-compartments, especially in mitochondria. S-glutathionylation and S-nitrosylation are modulated by fluctuations in reactive oxygen and nitrogen species and, in turn, orchestrate mitochondrial bioenergetics machinery, morphology, nutrients metabolism and apoptosis. In many neurodegenerative disorders, mitochondria dysfunction and oxidative/nitrosative stresses trigger or exacerbate their pathologies. Despite the substantial amount of research for most of these disorders, there are no successful treatments, while antioxidant supplementation failed in the majority of clinical trials. Herein, we discuss how S-glutathionylation and S-nitrosylation interfere in mitochondrial homeostasis and how the deregulation of these modifications is associated with Alzheimer’s, Parkinson’s, amyotrophic lateral sclerosis and Friedreich’s ataxia.
Collapse
Affiliation(s)
- Sofia Vrettou
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Rare Diseases, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
14
|
Discovery of Therapeutics Targeting Oxidative Stress in Autosomal Recessive Cerebellar Ataxia: A Systematic Review. Pharmaceuticals (Basel) 2022; 15:ph15060764. [PMID: 35745683 PMCID: PMC9228961 DOI: 10.3390/ph15060764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/05/2022] [Accepted: 06/14/2022] [Indexed: 01/05/2023] Open
Abstract
Autosomal recessive cerebellar ataxias (ARCAs) are a heterogeneous group of rare neurodegenerative inherited disorders. The resulting motor incoordination and progressive functional disabilities lead to reduced lifespan. There is currently no cure for ARCAs, likely attributed to the lack of understanding of the multifaceted roles of antioxidant defense and the underlying mechanisms. This systematic review aims to evaluate the extant literature on the current developments of therapeutic strategies that target oxidative stress for the management of ARCAs. We searched PubMed, Web of Science, and Science Direct Scopus for relevant peer-reviewed articles published from 1 January 2016 onwards. A total of 28 preclinical studies fulfilled the eligibility criteria for inclusion in this systematic review. We first evaluated the altered cellular processes, abnormal signaling cascades, and disrupted protein quality control underlying the pathogenesis of ARCA. We then examined the current potential therapeutic strategies for ARCAs, including aromatic, organic and pharmacological compounds, gene therapy, natural products, and nanotechnology, as well as their associated antioxidant pathways and modes of action. We then discussed their potential as antioxidant therapeutics for ARCAs, with the long-term view toward their possible translation to clinical practice. In conclusion, our current understanding is that these antioxidant therapies show promise in improving or halting the progression of ARCAs. Tailoring the therapies to specific disease stages could greatly facilitate the management of ARCAs.
Collapse
|
15
|
Abstract
INTRODUCTION Friedreich ataxia (FRDA) is an autosomal recessive disorder caused by deficiency of frataxin, an essential mitochondrial protein involved in iron sulfur cluster biogenesis, oxidative phosphorylation and other processes. FRDA most notably affects the heart, sensory neurons, spinal cord, cerebellum, and other brain regions, and manifests clinically as ataxia, sensory loss, dysarthria, spasticity, and hypertrophic cardiomyopathy. Therapeutic approaches in FRDA have consisted of two different approaches: (1) augmenting or restoring frataxin production and (2) modulating a variety of downstream processes related to mitochondrial dysfunction, including reactive oxygen species production, ferroptosis, or Nrf2 activation. AREAS COVERED In this review, we summarize data from major phase II clinical trials in FRDA published between 2015 and 2020, which includes A0001/EPI743, Omaveloxolone, RT001, and Actimmune. EXPERT OPINION A growing number of drug candidates are being tested in phase II clinical trials for FRDA; however, most have not met their primary endpoints, and none have received FDA approval. In this review, we aim to summarize completed phase II clinical trials in FRDA, outlining critical lessons that have been learned and that should be incorporated into future trial design to ultimately optimize drug development in FRDA.
Collapse
|
16
|
Melentev PA, Ryabova EV, Surina NV, Zhmujdina DR, Komissarov AE, Ivanova EA, Boltneva NP, Makhaeva GF, Sliusarenko MI, Yatsenko AS, Mohylyak II, Matiytsiv NP, Shcherbata HR, Sarantseva SV. Loss of swiss cheese in Neurons Contributes to Neurodegeneration with Mitochondria Abnormalities, Reactive Oxygen Species Acceleration and Accumulation of Lipid Droplets in Drosophila Brain. Int J Mol Sci 2021; 22:8275. [PMID: 34361042 PMCID: PMC8347196 DOI: 10.3390/ijms22158275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/17/2022] Open
Abstract
Various neurodegenerative disorders are associated with human NTE/PNPLA6 dysfunction. Mechanisms of neuropathogenesis in these diseases are far from clearly elucidated. Hereditary spastic paraplegia belongs to a type of neurodegeneration associated with NTE/PNLPLA6 and is implicated in neuron death. In this study, we used Drosophila melanogaster to investigate the consequences of neuronal knockdown of swiss cheese (sws)-the evolutionarily conserved ortholog of human NTE/PNPLA6-in vivo. Adult flies with the knockdown show longevity decline, locomotor and memory deficits, severe neurodegeneration progression in the brain, reactive oxygen species level acceleration, mitochondria abnormalities and lipid droplet accumulation. Our results suggest that SWS/NTE/PNPLA6 dysfunction in neurons induces oxidative stress and lipid metabolism alterations, involving mitochondria dynamics and lipid droplet turnover in neurodegeneration pathogenesis. We propose that there is a complex mechanism in neurological diseases such as hereditary spastic paraplegia, which includes a stress reaction, engaging mitochondria, lipid droplets and endoplasmic reticulum interplay.
Collapse
Affiliation(s)
- Pavel A. Melentev
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of NRC «Kurchatov Institute», 188300 Gatchina, Russia; (P.A.M.); (E.V.R.); (N.V.S.); (D.R.Z.); (A.E.K.); (E.A.I.)
| | - Elena V. Ryabova
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of NRC «Kurchatov Institute», 188300 Gatchina, Russia; (P.A.M.); (E.V.R.); (N.V.S.); (D.R.Z.); (A.E.K.); (E.A.I.)
| | - Nina V. Surina
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of NRC «Kurchatov Institute», 188300 Gatchina, Russia; (P.A.M.); (E.V.R.); (N.V.S.); (D.R.Z.); (A.E.K.); (E.A.I.)
| | - Darya R. Zhmujdina
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of NRC «Kurchatov Institute», 188300 Gatchina, Russia; (P.A.M.); (E.V.R.); (N.V.S.); (D.R.Z.); (A.E.K.); (E.A.I.)
| | - Artem E. Komissarov
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of NRC «Kurchatov Institute», 188300 Gatchina, Russia; (P.A.M.); (E.V.R.); (N.V.S.); (D.R.Z.); (A.E.K.); (E.A.I.)
| | - Ekaterina A. Ivanova
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of NRC «Kurchatov Institute», 188300 Gatchina, Russia; (P.A.M.); (E.V.R.); (N.V.S.); (D.R.Z.); (A.E.K.); (E.A.I.)
| | - Natalia P. Boltneva
- Institute of Physiologically Active Compounds Russian Academy of Sciences, 142432 Chernogolovka, Russia; (N.P.B.); (G.F.M.)
| | - Galina F. Makhaeva
- Institute of Physiologically Active Compounds Russian Academy of Sciences, 142432 Chernogolovka, Russia; (N.P.B.); (G.F.M.)
| | - Mariana I. Sliusarenko
- Institute of Cell Biochemistry, Hannover Medical School, 30625 Hannover, Germany; (M.I.S.); (A.S.Y.); (H.R.S.)
| | - Andriy S. Yatsenko
- Institute of Cell Biochemistry, Hannover Medical School, 30625 Hannover, Germany; (M.I.S.); (A.S.Y.); (H.R.S.)
| | - Iryna I. Mohylyak
- Department of Genetics and Biotechnology, Ivan Franko National University of Lviv, 79005 Lviv, Ukraine; (I.I.M.); (N.P.M.)
| | - Nataliya P. Matiytsiv
- Department of Genetics and Biotechnology, Ivan Franko National University of Lviv, 79005 Lviv, Ukraine; (I.I.M.); (N.P.M.)
| | - Halyna R. Shcherbata
- Institute of Cell Biochemistry, Hannover Medical School, 30625 Hannover, Germany; (M.I.S.); (A.S.Y.); (H.R.S.)
| | - Svetlana V. Sarantseva
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of NRC «Kurchatov Institute», 188300 Gatchina, Russia; (P.A.M.); (E.V.R.); (N.V.S.); (D.R.Z.); (A.E.K.); (E.A.I.)
| |
Collapse
|
17
|
Giustarini D, Milzani A, Dalle-Donne I, Rossi R. Measurement of S-glutathionylated proteins by HPLC. Amino Acids 2021; 54:675-686. [PMID: 34129091 PMCID: PMC9117368 DOI: 10.1007/s00726-021-03015-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/29/2021] [Indexed: 11/28/2022]
Abstract
S-glutathionylated proteins (GSSP), i.e., protein-mixed disulfides with glutathione (GSH), are considered a suitable biomarker of oxidative stress. In fact, they occur within cells at low level and their concentration increases markedly under pro-oxidant conditions. Plasma is something different, since it is physiologically rich in S-thiolated proteins (RSSP), i.e., protein-mixed disulfides with various types of low molecular mass thiols (LMM-SH). However, albumin, which is largely the most abundant plasma protein, possesses a cysteine residue at position 34 that is mostly reduced (about 60%) under physiological conditions, but easily involved in the formation of additional RSSP in the presence of oxidants. The quantification of GSSP requires special attention to sample handling, since their level can be overestimated as a result of artefactual oxidation of GSH. We have developed the present protocol to avoid this methodological problem. Samples should be treated as soon as possible after their collection with the alkylating agent N-ethylmaleimide that masks –SH groups and prevents their oxidation. The GSH released from mixed disulfides by reduction with dithiothreitol is then labeled with the fluorescent probe monobromobimane and quantified by HPLC. The method can be applied to many different biological samples, comprising blood components, red blood cell plasma membrane, cultured cells, and solid organs from animal models.
Collapse
Affiliation(s)
- Daniela Giustarini
- Department of Biotechnology, Chemistry and Pharmacy (Department of Excellence 2018-2022), Laboratory of Pharmacology and Toxicology, University of Siena, Via A. Moro 4, 53100, Siena, Italy.
| | - Aldo Milzani
- Department of Biosciences (Department of Excellence 2018-2022), Università Degli Studi Di Milano, via Celoria 26, I-20133, Milan, Italy
| | - Isabella Dalle-Donne
- Department of Biosciences (Department of Excellence 2018-2022), Università Degli Studi Di Milano, via Celoria 26, I-20133, Milan, Italy
| | - Ranieri Rossi
- Department of Biotechnology, Chemistry and Pharmacy (Department of Excellence 2018-2022), Laboratory of Pharmacology and Toxicology, University of Siena, Via A. Moro 4, 53100, Siena, Italy
| |
Collapse
|
18
|
Rubino FM. The Redox Potential of the β- 93-Cysteine Thiol Group in Human Hemoglobin Estimated from In Vitro Oxidant Challenge Experiments. Molecules 2021; 26:molecules26092528. [PMID: 33926119 PMCID: PMC8123695 DOI: 10.3390/molecules26092528] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 11/16/2022] Open
Abstract
Glutathionyl hemoglobin is a minor form of hemoglobin with intriguing properties. The measurement of the redox potential of its reactive β-93-Cysteine is useful to improve understanding of the response of erythrocytes to transient and chronic conditions of oxidative stress, where the level of glutathionyl hemoglobin is increased. An independent literature experiment describes the recovery of human erythrocytes exposed to an oxidant burst by measuring glutathione, glutathione disulfide and glutathionyl hemoglobin in a two-hour period. This article calculates a value for the redox potential E0 of the β-93-Cysteine, considering the erythrocyte as a closed system at equilibrium described by the Nernst equation and using the measurements of the literature experiment. The obtained value of E0 of −121 mV at pH 7.4 places hemoglobin as the most oxidizing thiol of the erythrocyte. By using as synthetic indicators of the concentrations the electrochemical potentials of the two main redox pairs in the erythrocytes, those of glutathione–glutathione disulfide and of glutathionyl–hemoglobin, the mechanism of the recovery phase can be hypothesized. Hemoglobin acts as the redox buffer that scavenges oxidized glutathione in the oxidative phase and releases it in the recovery phase, by acting as the substrate of the NAD(P)H-cofactored enzymes.
Collapse
Affiliation(s)
- Federico Maria Rubino
- LaTMA Laboratory for Analytical Toxicology and Metabonomics, Department of Health Sciences, Università degli Studi di Milano at "Ospedale San Paolo" v. A. di Rudinì 8, I-20142 Milano, Italy
| |
Collapse
|
19
|
Rebelo AP, Eidhof I, Cintra VP, Guillot-Noel L, Pereira CV, Timmann D, Traschütz A, Schöls L, Coarelli G, Durr A, Anheim M, Tranchant C, van de Warrenburg B, Guissart C, Koenig M, Howell J, Moraes CT, Schenck A, Stevanin G, Züchner S, Synofzik M. Biallelic loss-of-function variations in PRDX3 cause cerebellar ataxia. Brain 2021; 144:1467-1481. [PMID: 33889951 DOI: 10.1093/brain/awab071] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/13/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Peroxiredoxin 3 (PRDX3) belongs to a superfamily of peroxidases that function as protective antioxidant enzymes. Among the six isoforms (PRDX1-PRDX6), PRDX3 is the only protein exclusively localized to the mitochondria, which are the main source of reactive oxygen species. Excessive levels of reactive oxygen species are harmful to cells, inducing mitochondrial dysfunction, DNA damage, lipid and protein oxidation and ultimately apoptosis. Neuronal cell damage induced by oxidative stress has been associated with numerous neurodegenerative disorders including Alzheimer's and Parkinson's diseases. Leveraging the large aggregation of genomic ataxia datasets from the PREPARE (Preparing for Therapies in Autosomal Recessive Ataxias) network, we identified recessive mutations in PRDX3 as the genetic cause of cerebellar ataxia in five unrelated families, providing further evidence for oxidative stress in the pathogenesis of neurodegeneration. The clinical presentation of individuals with PRDX3 mutations consists of mild-to-moderate progressive cerebellar ataxia with concomitant hyper- and hypokinetic movement disorders, severe early-onset cerebellar atrophy, and in part olivary and brainstem degeneration. Patient fibroblasts showed a lack of PRDX3 protein, resulting in decreased glutathione peroxidase activity and decreased mitochondrial maximal respiratory capacity. Moreover, PRDX3 knockdown in cerebellar medulloblastoma cells resulted in significantly decreased cell viability, increased H2O2 levels and increased susceptibility to apoptosis triggered by reactive oxygen species. Pan-neuronal and pan-glial in vivo models of Drosophila revealed aberrant locomotor phenotypes and reduced survival times upon exposure to oxidative stress. Our findings reveal a central role for mitochondria and the implication of oxidative stress in PRDX3 disease pathogenesis and cerebellar vulnerability and suggest targets for future therapeutic approaches.
Collapse
Affiliation(s)
- Adriana P Rebelo
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, USA
| | - Ilse Eidhof
- Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Vivian P Cintra
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, USA
| | - Léna Guillot-Noel
- Sorbonne Université, Paris Brain Institute, AP-HP, INSERM, CNRS, Pitié-Salpêtrière University Hospital, Paris, France.,Neurogenetics Team, EPHE, PSL University, Paris, France
| | - Claudia V Pereira
- Departments of Neurology and Cell Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dagmar Timmann
- Department of Neurology, Essen University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Andreas Traschütz
- Translational Genomics of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), University of Tübingen, Tübingen, Germany
| | - Ludger Schöls
- Translational Genomics of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), University of Tübingen, Tübingen, Germany
| | - Giulia Coarelli
- Sorbonne Université, Paris Brain Institute, AP-HP, INSERM, CNRS, Pitié-Salpêtrière University Hospital, Paris, France
| | - Alexandra Durr
- Sorbonne Université, Paris Brain Institute, AP-HP, INSERM, CNRS, Pitié-Salpêtrière University Hospital, Paris, France.,Department of genetics, Hôpital de La Pitié-Salpétrière, Paris, France
| | - Mathieu Anheim
- Département de Neurologie, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.,Institute of Genetics and Molecular and Cellular Biology, INSERM-U964/CNRS-UMR7104, University of Strasbourg, Illkirch, France
| | - Christine Tranchant
- Département de Neurologie, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France.,Institute of Genetics and Molecular and Cellular Biology, INSERM-U964/CNRS-UMR7104, University of Strasbourg, Illkirch, France
| | - Bart van de Warrenburg
- Department of Neurology, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Claire Guissart
- EA7402 Institut Universitaire de Recherche Clinique and Laboratoire de Génétique Moléculaire, CHU and Université de Montpellier, Montpellier, France
| | - Michel Koenig
- EA7402 Institut Universitaire de Recherche Clinique and Laboratoire de Génétique Moléculaire, CHU and Université de Montpellier, Montpellier, France
| | - Jack Howell
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, USA
| | - Carlos T Moraes
- Departments of Neurology and Cell Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Giovanni Stevanin
- Sorbonne Université, Paris Brain Institute, AP-HP, INSERM, CNRS, Pitié-Salpêtrière University Hospital, Paris, France.,Neurogenetics Team, EPHE, PSL University, Paris, France
| | - Stephan Züchner
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, USA
| | - Matthis Synofzik
- Translational Genomics of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), University of Tübingen, Tübingen, Germany
| | | |
Collapse
|
20
|
Pallardó FV, Pagano G, Rodríguez LR, Gonzalez-Cabo P, Lyakhovich A, Trifuoggi M. Friedreich Ataxia: current state-of-the-art, and future prospects for mitochondrial-focused therapies. Transl Res 2021; 229:135-141. [PMID: 32841735 DOI: 10.1016/j.trsl.2020.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022]
Abstract
Friedreich's Ataxia is an autosomal recessive genetic disease causing the defective gene product, frataxin. A body of literature has been focused on the attempts to counteract frataxin deficiency and the consequent iron imbalance, in order to mitigate the disease-associated pro-oxidant state and clinical course. The present mini review is aimed at evaluating the basic and clinical reports on the roles and the use of a set of iron chelators, antioxidants and some cofactors involved in the key mitochondrial functions. Extensive literature has focused on the protective roles of iron chelators, coenzyme Q10 and analogs, and vitamin E, altogether with varying outcomes in clinical studies. Other studies have suggested mitoprotective roles for other mitochondrial cofactors, involved in Krebs cycle, such as alpha-lipoic acid and carnitine, involved in acyl transport across the mitochondrial membrane. A body of evidence points to the strong antioxidant properties of these cofactors, and to their potential contribution in mitoprotective strategies in Friedreich's Ataxia clinical evolution. Thus, we suggest the rationale for planning combination strategies based on the 3 mitochondrial cofactors and of some antioxidants and iron binders as mitoprotective cocktails in Friedreich Ataxia patients, calling attention to clinical practitioners of the importance to implement clinical trials.
Collapse
Affiliation(s)
- Federico V Pallardó
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-INCLIVA, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain.
| | - Giovanni Pagano
- Department of Chemical Sciences, Federico II Naples University, Naples, Italy
| | - Laura R Rodríguez
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-INCLIVA, Valencia, Spain
| | - Pilar Gonzalez-Cabo
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-INCLIVA, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Alex Lyakhovich
- Vall d'Hebron Institut de Recerca, Barcelona, Catalunya, Spain; Institute of Molecular Biology and Biophysics of the "Federal Research Center of Fundamental and Translational Medicine", Novosibirsk, Russia
| | - Marco Trifuoggi
- Department of Chemical Sciences, Federico II Naples University, Naples, Italy
| |
Collapse
|
21
|
Petrillo S, Santoro M, La Rosa P, Perna A, Gallo MG, Bertini ES, Silvestri G, Piemonte F. Nuclear Factor Erythroid 2-Related Factor 2 Activation Might Mitigate Clinical Symptoms in Friedreich's Ataxia: Clues of an "Out-Brain Origin" of the Disease From a Family Study. Front Neurosci 2021; 15:638810. [PMID: 33708070 PMCID: PMC7940825 DOI: 10.3389/fnins.2021.638810] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/22/2021] [Indexed: 11/25/2022] Open
Abstract
Friedreich’s ataxia (FRDA) is the most frequent autosomal recessive ataxia in western countries, with a mean age of onset at 10–15 years. Patients manifest progressive cerebellar and sensory ataxia, dysarthria, lower limb pyramidal weakness, and other systemic manifestations. Previously, we described a family displaying two expanded GAA alleles not only in the proband affected by late-onset FRDA but also in the two asymptomatic family members: the mother and the younger sister. Both of them showed a significant reduction of frataxin levels, without any disease manifestation. Here, we analyzed if a protective mechanism might contribute to modulate the phenotype in this family. We particularly focused on the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2), the first line of antioxidant defense in cells, and on the glutathione (GSH) system, an index of reactive oxygen species (ROS) detoxification ability. Our findings show a great reactivity of the GSH system to the frataxin deficiency, particularly in the asymptomatic mother, where the genes of GSH synthesis [glutamate–cysteine ligase (GCL)] and GSSG detoxification [GSH S-reductase (GSR)] were highly responsive. The GSR was activated even in the asymptomatic sister and in the proband, reflecting the need of buffering the GSSG increase. Furthermore, and contrasting the NRF2 expression documented in FRDA tissues, NRF2 was highly activated in the mother and in the younger sister, while it was constitutively low in the proband. This suggests that, also under frataxin depletion, the endogenous stimulation of NRF2 in asymptomatic FRDA subjects may contribute to protect against the progressive oxidative damage, helping to prevent the onset of neurological symptoms and highlighting an “out-brain origin” of the disease.
Collapse
Affiliation(s)
- Sara Petrillo
- Unit of Muscular and Neurodegenerative Diseases, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | | | - Piergiorgio La Rosa
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Alessia Perna
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Giovanna Gallo
- Unit of Muscular and Neurodegenerative Diseases, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Enrico Silvio Bertini
- Unit of Muscular and Neurodegenerative Diseases, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Gabriella Silvestri
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Rome, Italy.,UOC of Neurology, Area of Neuroscience, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Fiorella Piemonte
- Unit of Muscular and Neurodegenerative Diseases, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| |
Collapse
|
22
|
Phang MWL, Lew SY, Chung I, Lim WKS, Lim LW, Wong KH. Therapeutic roles of natural remedies in combating hereditary ataxia: A systematic review. Chin Med 2021; 16:15. [PMID: 33509239 PMCID: PMC7841890 DOI: 10.1186/s13020-020-00414-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/17/2020] [Accepted: 12/11/2020] [Indexed: 12/30/2022] Open
Abstract
Background Hereditary ataxia (HA) represents a group of genetically heterogeneous neurodegenerative diseases caused by dysfunction of the cerebellum or disruption of the connection between the cerebellum and other areas of the central nervous system. Phenotypic manifestation of HA includes unsteadiness of stance and gait, dysarthria, nystagmus, dysmetria and complaints of clumsiness. There are no specific treatments for HA. Management strategies provide supportive treatment to reduce symptoms. Objectives This systematic review aimed to identify, evaluate and summarise the published literature on the therapeutic roles of natural remedies in the treatment of HA to provide evidence for clinical practice. Methods A systematic literature search was conducted using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). Web of Science, PubMed and Science Direct Scopus were thoroughly searched for relevant published articles from June 2007 to July 2020. Results Ten pre-clinical and two clinical studies were eligible for inclusion in this systematic review. We identified the therapeutic roles of medicinal plants Brassica napus, Gardenia jasminoides, Gastrodia elata, Ginkgo biloba, Glycyrrhiza inflata, Paeonia lactiflora, Pueraria lobata and Rehmannia glutinosa; herbal formulations Shaoyao Gancao Tang and Zhengan Xifeng Tang; and medicinal mushroom Hericium erinaceus in the treatment of HA. In this review, we evaluated the mode of actions contributing to their therapeutic effects, including activation of the ubiquitin–proteasome system, activation of antioxidant pathways, maintenance of intracellular calcium homeostasis and regulation of chaperones. We also briefly highlighted the integral cellular signalling pathways responsible for orchestrating the mode of actions. Conclusion We reviewed the therapeutic roles of natural remedies in improving or halting the progression of HA, which warrant further study for applications into clinical practice.
Collapse
Affiliation(s)
- Michael Weng Lok Phang
- Department of Anatomy, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Sze Yuen Lew
- Department of Anatomy, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Ivy Chung
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - William Kiong-Seng Lim
- Faculty of Medicine and Health Sciences, Universiti Malaysia Sarawak, Kuching, Sarawak, 94300, Malaysia
| | - Lee Wei Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong Special Administrative Region, China.
| | - Kah Hui Wong
- Department of Anatomy, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| |
Collapse
|
23
|
Seco-Cervera M, González-Cabo P, Pallardó FV, Romá-Mateo C, García-Giménez JL. Thioredoxin and Glutaredoxin Systems as Potential Targets for the Development of New Treatments in Friedreich's Ataxia. Antioxidants (Basel) 2020; 9:antiox9121257. [PMID: 33321938 PMCID: PMC7763308 DOI: 10.3390/antiox9121257] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
The thioredoxin family consists of a small group of redox proteins present in all organisms and composed of thioredoxins (TRXs), glutaredoxins (GLRXs) and peroxiredoxins (PRDXs) which are found in the extracellular fluid, the cytoplasm, the mitochondria and in the nucleus with functions that include antioxidation, signaling and transcriptional control, among others. The importance of thioredoxin family proteins in neurodegenerative diseases is gaining relevance because some of these proteins have demonstrated an important role in the central nervous system by mediating neuroprotection against oxidative stress, contributing to mitochondrial function and regulating gene expression. Specifically, in the context of Friedreich’s ataxia (FRDA), thioredoxin family proteins may have a special role in the regulation of Nrf2 expression and function, in Fe-S cluster metabolism, controlling the expression of genes located at the iron-response element (IRE) and probably regulating ferroptosis. Therefore, comprehension of the mechanisms that closely link thioredoxin family proteins with cellular processes affected in FRDA will serve as a cornerstone to design improved therapeutic strategies.
Collapse
Affiliation(s)
- Marta Seco-Cervera
- Centre for Biomedical Research on Rare Diseases (CIBERER), 46010 Valencia, Spain; (M.S.-C.); (P.G.-C.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València (UV), 46010 Valencia, Spain
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
| | - Pilar González-Cabo
- Centre for Biomedical Research on Rare Diseases (CIBERER), 46010 Valencia, Spain; (M.S.-C.); (P.G.-C.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València (UV), 46010 Valencia, Spain
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
| | - Federico V. Pallardó
- Centre for Biomedical Research on Rare Diseases (CIBERER), 46010 Valencia, Spain; (M.S.-C.); (P.G.-C.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València (UV), 46010 Valencia, Spain
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
| | - Carlos Romá-Mateo
- Centre for Biomedical Research on Rare Diseases (CIBERER), 46010 Valencia, Spain; (M.S.-C.); (P.G.-C.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València (UV), 46010 Valencia, Spain
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Correspondence: (C.R.-M.); (J.L.G.-G.); Tel.: +34-963-864-646 (C.R.-M. & J.L.G.-G.)
| | - José Luis García-Giménez
- Centre for Biomedical Research on Rare Diseases (CIBERER), 46010 Valencia, Spain; (M.S.-C.); (P.G.-C.); (F.V.P.)
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València (UV), 46010 Valencia, Spain
- Biomedical Research Institute INCLIVA, 46010 Valencia, Spain
- Correspondence: (C.R.-M.); (J.L.G.-G.); Tel.: +34-963-864-646 (C.R.-M. & J.L.G.-G.)
| |
Collapse
|
24
|
La Rosa P, Petrillo S, Fiorenza MT, Bertini ES, Piemonte F. Ferroptosis in Friedreich's Ataxia: A Metal-Induced Neurodegenerative Disease. Biomolecules 2020; 10:biom10111551. [PMID: 33202971 PMCID: PMC7696618 DOI: 10.3390/biom10111551] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
Ferroptosis is an iron-dependent form of regulated cell death, arising from the accumulation of lipid-based reactive oxygen species when glutathione-dependent repair systems are compromised. Lipid peroxidation, mitochondrial impairment and iron dyshomeostasis are the hallmark of ferroptosis, which is emerging as a crucial player in neurodegeneration. This review provides an analysis of the most recent advances in ferroptosis, with a special focus on Friedreich's Ataxia (FA), the most common autosomal recessive neurodegenerative disease, caused by reduced levels of frataxin, a mitochondrial protein involved in iron-sulfur cluster synthesis and antioxidant defenses. The hypothesis is that the iron-induced oxidative damage accumulates over time in FA, lowering the ferroptosis threshold and leading to neuronal cell death and, at last, to cardiac failure. The use of anti-ferroptosis drugs combined with treatments able to activate the antioxidant response will be of paramount importance in FA therapy, such as in many other neurodegenerative diseases triggered by oxidative stress.
Collapse
Affiliation(s)
- Piergiorgio La Rosa
- Department of Psychology, Division of Neuroscience, Sapienza University of Rome, 00185 Rome, Italy; (P.L.R.); (M.T.F.)
| | - Sara Petrillo
- Unit of Muscular and Neurodegenerative Diseases, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (E.S.B.)
| | - Maria Teresa Fiorenza
- Department of Psychology, Division of Neuroscience, Sapienza University of Rome, 00185 Rome, Italy; (P.L.R.); (M.T.F.)
| | - Enrico Silvio Bertini
- Unit of Muscular and Neurodegenerative Diseases, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (E.S.B.)
| | - Fiorella Piemonte
- Unit of Muscular and Neurodegenerative Diseases, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (E.S.B.)
- Correspondence: ; Tel.: +39-06-6859-2102
| |
Collapse
|
25
|
Smith FM, Kosman DJ. Molecular Defects in Friedreich's Ataxia: Convergence of Oxidative Stress and Cytoskeletal Abnormalities. Front Mol Biosci 2020; 7:569293. [PMID: 33263002 PMCID: PMC7686857 DOI: 10.3389/fmolb.2020.569293] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/10/2020] [Indexed: 01/18/2023] Open
Abstract
Friedreich’s ataxia (FRDA) is a multi-faceted disease characterized by progressive sensory–motor loss, neurodegeneration, brain iron accumulation, and eventual death by hypertrophic cardiomyopathy. FRDA follows loss of frataxin (FXN), a mitochondrial chaperone protein required for incorporation of iron into iron–sulfur cluster and heme precursors. After the discovery of the molecular basis of FRDA in 1996, over two decades of research have been dedicated to understanding the temporal manifestations of disease both at the whole body and molecular level. Early research indicated strong cellular iron dysregulation in both human and yeast models followed by onset of oxidative stress. Since then, the pathophysiology due to dysregulation of intracellular iron chaperoning has become central in FRDA relative to antioxidant defense and run-down in energy metabolism. At the same time, limited consideration has been given to changes in cytoskeletal organization, which was one of the first molecular defects noted. These alterations include both post-translational oxidative glutathionylation of actin monomers and differential DNA processing of a cytoskeletal regulator PIP5K1β. Currently unknown in respect to FRDA but well understood in the context of FXN-deficient cell physiology is the resulting impact on the cytoskeleton; this disassembly of actin filaments has a particularly profound effect on cell–cell junctions characteristic of barrier cells. With respect to a neurodegenerative disorder such as FRDA, this cytoskeletal and tight junction breakdown in the brain microvascular endothelial cells of the blood–brain barrier is likely a component of disease etiology. This review serves to outline a brief history of this research and hones in on pathway dysregulation downstream of iron-related pathology in FRDA related to actin dynamics. The review presented here was not written with the intent of being exhaustive, but to instead urge the reader to consider the essentiality of the cytoskeleton and appreciate the limited knowledge on FRDA-related cytoskeletal dysfunction as a result of oxidative stress. The review examines previous hypotheses of neurodegeneration with brain iron accumulation (NBIA) in FRDA with a specific biochemical focus.
Collapse
Affiliation(s)
- Frances M Smith
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, United States
| | - Daniel J Kosman
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
26
|
Chiang S, Huang MLH, Richardson DR. Treatment of dilated cardiomyopathy in a mouse model of Friedreich's ataxia using N-acetylcysteine and identification of alterations in microRNA expression that could be involved in its pathogenesis. Pharmacol Res 2020; 159:104994. [PMID: 32534099 DOI: 10.1016/j.phrs.2020.104994] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/01/2020] [Accepted: 06/01/2020] [Indexed: 01/01/2023]
Abstract
Deficient expression of the mitochondrial protein, frataxin, leads to a deadly cardiomyopathy. Our laboratory reported the master regulator of oxidative stress, nuclear factor erythroid 2-related factor-2 (Nrf2), demonstrates marked down-regulation after frataxin deletion in the heart. This was due, in part, to a pronounced increase in Keap1. To assess if this can be therapeutically targeted, cells were incubated with N-acetylcysteine (NAC), or buthionine sulfoximine (BSO), which increases or decreases glutathione (GSH), respectively, or the NRF2-inducer, sulforaphane (SFN). While SFN significantly (p < 0.05) induced NRF2, KEAP1 and BACH1, NAC attenuated SFN-induced NRF2, KEAP1 and BACH1. The down-regulation of KEAP1 by NAC was of interest, as Keap1 is markedly increased in the MCK conditional frataxin knockout (MCK KO) mouse model and this could lead to the decreased Nrf2 levels. Considering this, MCK KO mice were treated with i.p. NAC (500- or 1500-mg/kg, 5 days/week for 5-weeks) and demonstrated slightly less (p > 0.05) body weight loss versus the vehicle-treated KO. However, NAC did not rescue the cardiomyopathy. To additionally examine the dys-regulation of Nrf2 upon frataxin deletion, studies assessed the role of microRNA (miRNA) in this process. In MCK KO mice, miR-144 was up-regulated, which down-regulates Nrf2. Furthermore, miRNA screening in MCK KO mice demonstrated 23 miRNAs from 756 screened were significantly (p < 0.05) altered in KOs versus WT littermates. Of these, miR-21*, miR-34c*, and miR-200c, demonstrated marked alterations, with functional clustering analysis showing they regulate genes linked to cardiac hypertrophy, cardiomyopathy, and oxidative stress, respectively.
Collapse
MESH Headings
- Acetylcysteine/pharmacology
- Animals
- Basic-Leucine Zipper Transcription Factors/metabolism
- Cardiomyopathy, Dilated/drug therapy
- Cardiomyopathy, Dilated/etiology
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cell Line, Tumor
- Disease Models, Animal
- Friedreich Ataxia/complications
- Friedreich Ataxia/genetics
- Gene Expression Regulation
- Humans
- Iron-Binding Proteins/genetics
- Iron-Binding Proteins/metabolism
- Isothiocyanates/pharmacology
- Kelch-Like ECH-Associated Protein 1/metabolism
- Mice, Knockout
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- NF-E2-Related Factor 2/genetics
- NF-E2-Related Factor 2/metabolism
- Sulfoxides/pharmacology
- Frataxin
Collapse
Affiliation(s)
- S Chiang
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building (K25), University of Sydney, Sydney, New South Wales, 2006 Australia
| | - M L H Huang
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building (K25), University of Sydney, Sydney, New South Wales, 2006 Australia
| | - D R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building (K25), University of Sydney, Sydney, New South Wales, 2006 Australia; Centre for Cancer Cell Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia.
| |
Collapse
|
27
|
Rodríguez LR, Lapeña T, Calap-Quintana P, Moltó MD, Gonzalez-Cabo P, Navarro Langa JA. Antioxidant Therapies and Oxidative Stress in Friedreich´s Ataxia: The Right Path or Just a Diversion? Antioxidants (Basel) 2020; 9:E664. [PMID: 32722309 PMCID: PMC7465446 DOI: 10.3390/antiox9080664] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/17/2020] [Accepted: 07/19/2020] [Indexed: 12/12/2022] Open
Abstract
Friedreich´s ataxia is the commonest autosomal recessive ataxia among population of European descent. Despite the huge advances performed in the last decades, a cure still remains elusive. One of the most studied hallmarks of the disease is the increased production of oxidative stress markers in patients and models. This feature has been the motivation to develop treatments that aim to counteract such boost of free radicals and to enhance the production of antioxidant defenses. In this work, we present and critically review those "antioxidant" drugs that went beyond the disease´s models and were approved for its application in clinical trials. The evaluation of these trials highlights some crucial aspects of the FRDA research. On the one hand, the analysis contributes to elucidate whether oxidative stress plays a central role or whether it is only an epiphenomenon. On the other hand, it comments on some limitations in the current trials that complicate the analysis and interpretation of their outcome. We also include some suggestions that will be interesting to implement in future studies and clinical trials.
Collapse
Affiliation(s)
- Laura R. Rodríguez
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València-INCLIVA, 46010 Valencia, Spain; (L.R.R.); (T.L.); (P.C.-Q.)
- Associated Unit for Rare Diseases INCLIVA-CIPF, 46010 Valencia, Spain
| | - Tamara Lapeña
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València-INCLIVA, 46010 Valencia, Spain; (L.R.R.); (T.L.); (P.C.-Q.)
- Associated Unit for Rare Diseases INCLIVA-CIPF, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | - Pablo Calap-Quintana
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València-INCLIVA, 46010 Valencia, Spain; (L.R.R.); (T.L.); (P.C.-Q.)
- Associated Unit for Rare Diseases INCLIVA-CIPF, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | - María Dolores Moltó
- Department of Genetics, Universitat de València-INCLIVA, 46100 Valencia, Spain;
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), 46100 Valencia, Spain
| | - Pilar Gonzalez-Cabo
- Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València-INCLIVA, 46010 Valencia, Spain; (L.R.R.); (T.L.); (P.C.-Q.)
- Associated Unit for Rare Diseases INCLIVA-CIPF, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | | |
Collapse
|
28
|
Purroy R, Medina-Carbonero M, Ros J, Tamarit J. Frataxin-deficient cardiomyocytes present an altered thiol-redox state which targets actin and pyruvate dehydrogenase. Redox Biol 2020; 32:101520. [PMID: 32279039 PMCID: PMC7152683 DOI: 10.1016/j.redox.2020.101520] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/10/2020] [Accepted: 03/21/2020] [Indexed: 01/11/2023] Open
Abstract
Friedreich ataxia (FA) is a cardioneurodegenerative disease caused by deficient frataxin expression. This mitochondrial protein has been related to iron homeostasis, energy metabolism, and oxidative stress. Previously, we set up a cardiac cellular model of FA based on neonatal rat cardiac myocytes (NRVM) and lentivirus-mediated frataxin RNA interference. These frataxin-deficient NRVMs presented lipid droplet accumulation, mitochondrial swelling and signs of oxidative stress. Therefore, we decided to explore the presence of protein thiol modifications in this model. With this purpose, reduced glutathione (GSH) levels were measured and the presence of glutathionylated proteins was analyzed. We observed decreased GSH content and increased presence of glutahionylated actin in frataxin-deficient NRVMs. Moreover, the presence of oxidized cysteine residues was investigated using the thiol-reactive fluorescent probe iodoacetamide-Bodipy and 2D-gel electrophoresis. With this approach, we identified two proteins with altered redox status in frataxin-deficient NRVMs: electron transfer flavoprotein-ubiquinone oxidoreductase and dihydrolipoyl dehydrogenase (DLDH). As DLDH is involved in protein-bound lipoic acid redox cycling, we analyzed the redox state of this cofactor and we observed that lipoic acid from pyruvate dehydrogenase was more oxidized in frataxin-deficient cells. Also, by targeted proteomics, we observed a decreased content on the PDH A1 subunit from pyruvate dehydrogenase. Finally, we analyzed the consequences of supplementing frataxin-deficient NRVMs with the PDH cofactors thiamine and lipoic acid, the PDH activator dichloroacetate and the antioxidants N-acetyl cysteine and Tiron. Both dichloroacetate and Tiron were able to partially prevent lipid droplet accumulation in these cells. Overall, these results indicate that frataxin-deficient NRVMs present an altered thiol-redox state which could contribute to the cardiac pathology.
Collapse
Affiliation(s)
- Rosa Purroy
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLLeida, Universitat de Lleida, Lleida, Spain
| | - Marta Medina-Carbonero
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLLeida, Universitat de Lleida, Lleida, Spain
| | - Joaquim Ros
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLLeida, Universitat de Lleida, Lleida, Spain
| | - Jordi Tamarit
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLLeida, Universitat de Lleida, Lleida, Spain.
| |
Collapse
|
29
|
Chen HJC, Lai PY, Wu DC. Analysis of cysteine glutathionylation in hemoglobin of gastric cancer patients using nanoflow liquid chromatography/triple-stage mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2020; 34 Suppl 1:e8588. [PMID: 31509281 DOI: 10.1002/rcm.8588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 09/03/2019] [Accepted: 09/03/2019] [Indexed: 06/10/2023]
Abstract
Glutathione is an intracellular antioxidant capable of scavenging free radicals and detoxifying electrophiles from endogenous and exogenous sources via the free thiol group. Post-translational glutathionylation at cysteine residues of proteins can affect the structure and cause a functional change of proteins. Protein glutathionylation has been proven to reflect the cellular redox status. Our previous report indicates that the levels of glutathionylation in hemoglobin from peripheral blood of smokers are significantly higher than in nonsmokers. In this study, a nanoflow liquid chromatography/nanospray ionization triple-stage mass spectrometric (nanoLC/NSI-MS3 ) method with a linear ion trap mass spectrometer was employed to quantify glutathionylated peptides in the trypsin digests of hemoglobin from gastric cancer patients. We compare the extent of glutathionylation in hemoglobin from nonsmoking gastric cancer patients with that from nonsmoking healthy adults. Using a carboxymethylated peptide as the reference peptide, the relative quantification of each glutathionylated peptide was measured as the peak area ratio of the modified peptide versus the sum of the peak areas of the modified and the carboxymethylated parent peptide in the selected reaction monitoring chromatograms. Using this method, we found that the extents of glutathionylation at Cys-104 of the α-globin and Cys-93 of β-globulin hemoglobin from 10 gastric cancer patients were significantly higher than those from 14 normal individuals with p values <0.0001. Our results suggest the possibility of using the extent of cysteine glutathionylation at β-93 of hemoglobin as an oxidative stress biomarker candidate for gastric cancer.
Collapse
Affiliation(s)
- Hauh-Jyun Candy Chen
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Ming-Hsiung, Chia-Yi, 62142, Taiwan
| | - Pang-Yen Lai
- Department of Chemistry and Biochemistry, National Chung Cheng University, 168 University Road, Ming-Hsiung, Chia-Yi, 62142, Taiwan
| | - Deng-Chyang Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
30
|
Bolotta A, Pini A, Abruzzo PM, Ghezzo A, Modesti A, Gamberi T, Ferreri C, Bugamelli F, Fortuna F, Vertuani S, Manfredini S, Zucchini C, Marini M. Effects of tocotrienol supplementation in Friedreich's ataxia: A model of oxidative stress pathology. Exp Biol Med (Maywood) 2020; 245:201-212. [PMID: 31795754 PMCID: PMC7045332 DOI: 10.1177/1535370219890873] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 11/04/2019] [Indexed: 01/08/2023] Open
Abstract
Friedreich’s ataxia is an autosomal recessive disorder characterized by impaired mitochondrial function, resulting in oxidative stress. In this study, we aimed at evaluating whether tocotrienol, a phytonutrient that diffuses easily in tissues with saturated fatty layers, could complement the current treatment with idebenone, a quinone analogue with antioxidant properties. Five young Friedreich’s ataxia patients received a low-dose tocotrienol supplementation (5 mg/kg/day), while not discontinuing idebenone treatment. Several oxidative stress markers and biological parameters related to oxidative stress were evaluated at the time of initiation of treatment and 2 and 12 months post-treatment. Some oxidative stress-related parameters and some inflammation indices were altered in Friedreich’s ataxia patients taking idebenone alone and tended to be normal values following tocotrienol supplementation; likewise, a cardiac magnetic resonance study showed some improvement following one-year tocotrienol treatment. The pathway by which tocotrienol affects the Nrf2 modulation of hepcidin gene expression, a peptide involved in iron handling and in inflammatory responses, is viewed in the light of the disruption of the iron intracellular distribution and of the Nrf2 anergy characterizing Friedreich’s ataxia. This research provides a suitable model to analyze the efficacy of therapeutic strategies able to counteract the excess free radicals in Friedreich’s ataxia, and paves the way to long-term clinical studies.
Collapse
Affiliation(s)
- Alessandra Bolotta
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Milan 20148, Italy
| | - Antonella Pini
- Child Neurology and Psychiatry Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna 40139, Italy
| | - Provvidenza M Abruzzo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Milan 20148, Italy
| | - Alessandro Ghezzo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy
| | - Alessandra Modesti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Firenze 50134, Italy
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Firenze 50134, Italy
| | | | - Francesca Bugamelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Filippo Fortuna
- Neurochemistry Laboratory, Azienda Ospedaliera Ospedali Riuniti Marche Nord, Pesaro 61121, Italy
| | - Silvia Vertuani
- Department of Pharmaceutical Sciences, University of Ferrara, Ferrara 44100, Italy
| | - Stefano Manfredini
- Department of Pharmaceutical Sciences, University of Ferrara, Ferrara 44100, Italy
| | - Cinzia Zucchini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy
| | - Marina Marini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40126, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Milan 20148, Italy
| |
Collapse
|
31
|
The NRF2 Signaling Network Defines Clinical Biomarkers and Therapeutic Opportunity in Friedreich's Ataxia. Int J Mol Sci 2020; 21:ijms21030916. [PMID: 32019240 PMCID: PMC7037688 DOI: 10.3390/ijms21030916] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/06/2023] Open
Abstract
Friedreich’s ataxia (FA) is a trinucleotide repeats expansion neurodegenerative disorder, for which no cure or approved therapies are present. In most cases, GAA trinucleotide repetitions in the first intron of the FXN gene are the genetic trigger of FA, determining a strong reduction of frataxin, a mitochondrial protein involved in iron homeostasis. Frataxin depletion impairs iron–sulfur cluster biosynthesis and determines iron accumulation in the mitochondria. Mounting evidence suggests that these defects increase oxidative stress susceptibility and reactive oxygen species production in FA, where the pathologic picture is worsened by a defective regulation of the expression and signaling pathway modulation of the transcription factor NF-E2 p45-related factor 2 (NRF2), one of the fundamental mediators of the cellular antioxidant response. NRF2 protein downregulation and impairment of its nuclear translocation can compromise the adequate cellular response to the frataxin depletion-dependent redox imbalance. As NRF2 stability, expression, and activation can be modulated by diverse natural and synthetic compounds, efforts have been made in recent years to understand if regulating NRF2 signaling might ameliorate the pathologic defects in FA. Here we provide an analysis of the pharmaceutical interventions aimed at restoring the NRF2 signaling network in FA, elucidating specific biomarkers useful for monitoring therapeutic effectiveness, and developing new therapeutic tools.
Collapse
|
32
|
Mathew B, Srinivasan K, Johnson P, Thomas T, Mandal AK. Elevated levels of glutathionyl haemoglobin as an oxidative stress marker in patients with major depressive disorder. Indian J Med Res 2020; 149:497-502. [PMID: 31411173 PMCID: PMC6676840 DOI: 10.4103/ijmr.ijmr_586_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Background & objectives: Oxidative stress has been implicated in the pathophysiology of major depressive disorder (MDD), but biomarkers to assess oxidative stress in patients with MDD have yielded ambiguous results. Glutathionyl haemoglobin (GS-Hb) has been reported as a stable and potential biomarker for oxidative stress in various clinical conditions. The objective of the study was to evaluate GS-Hb as a potential biomarker of oxidative stress in patients with MDD through its quantification and to compare the levels of GS-Hb in age- and gender-matched healthy controls. Methods: The levels of GS-Hb were estimated using liquid chromatography coupled to electrospray ionization mass spectrometry in patients diagnosed with MDD and in a subset of patients after six weeks of treatment with selective serotonin reuptake inhibitors (SSRIs). Results: GS-Hb levels in drug-naïve patients with MDD (n=26) were significantly elevated compared to matched healthy controls (n=17). GS-Hb levels were not significantly different between MDD patients with and without co-morbid anxiety disorders. There were no significant differences in GS-Hb levels following six weeks of treatment with SSRIs compared to baseline. Interpretation & conclusions: Compared to controls, GS-Hb level in patients with MDD was significantly elevated, suggestive of increased oxidative stress associated with MDD. However, six weeks of antidepressant treatment was not sufficient to modify the alterations in antioxidant/oxidant system. Further studies need to be done with a large sample of MDD patients with a longer duration of antidepressant treatment.
Collapse
Affiliation(s)
- Boby Mathew
- Clinical Proteomics Unit, Division of Molecular Medicine, St. John's Research Institute, Bengaluru, India
| | - Krishnamachari Srinivasan
- Department of Psychiatry, St. John's Medical College & Hospital, St. John's National Academy of Health Sciences, Bengaluru, India
| | - Pradeep Johnson
- Department of Psychiatry, St. John's Medical College & Hospital, St. John's National Academy of Health Sciences, Bengaluru, India
| | - Tinku Thomas
- Division of Epidemiology & Biostatistics, St. John's Research Institute, Bengaluru, India
| | - Amit Kumar Mandal
- Clinical Proteomics Unit, Division of Molecular Medicine, St. John's Research Institute, Bengaluru, India
| |
Collapse
|
33
|
McMackin MZ, Durbin-Johnson B, Napierala M, Napierala JS, Ruiz L, Napoli E, Perlman S, Giulivi C, Cortopassi GA. Potential biomarker identification for Friedreich's ataxia using overlapping gene expression patterns in patient cells and mouse dorsal root ganglion. PLoS One 2019; 14:e0223209. [PMID: 31665133 PMCID: PMC6821053 DOI: 10.1371/journal.pone.0223209] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/15/2019] [Indexed: 12/22/2022] Open
Abstract
Friedreich's ataxia (FA) is a neurodegenerative disease with no approved therapy that is the result of frataxin deficiency. The identification of human FA blood biomarkers related to disease severity and neuro-pathomechanism could support clinical trials of drug efficacy. To try to identify human biomarkers of neuro-pathomechanistic relevance, we compared the overlapping gene expression changes of primary blood and skin cells of FA patients with changes in the Dorsal Root Ganglion (DRG) of the KIKO FA mouse model. As DRG is the primary site of neurodegeneration in FA, our goal was to identify which changes in blood and skin of FA patients provide a 'window' into the FA neuropathomechanism inside the nervous system. In addition, gene expression in frataxin-deficient neuroglial cells and FA mouse hearts were compared for a total of 5 data sets. The overlap of these changes strongly supports mitochondrial changes, apoptosis and alterations of selenium metabolism. Consistent biomarkers were observed, including three genes of mitochondrial stress (MTIF2, ENO2), apoptosis (DDIT3/CHOP), oxidative stress (PREX1), and selenometabolism (SEPW1). These results prompted our investigation of the GPX1 activity as a marker of selenium and oxidative stress, in which we observed a significant change in FA patients. We believe these lead biomarkers that could be assayed in FA patient blood as indicators of disease severity and progression, and also support the involvement of mitochondria, apoptosis and selenium in the neurodegenerative process.
Collapse
Affiliation(s)
- Marissa Z. McMackin
- Department of Molecular Biosciences, University of California, Davis, Davis, California, United States of America
| | - Blythe Durbin-Johnson
- Bioinformatics, University of California, Davis, Davis, California, United States of America
| | - Marek Napierala
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jill S. Napierala
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Luis Ruiz
- Department of Molecular Biosciences, University of California, Davis, Davis, California, United States of America
| | - Eleonora Napoli
- Department of Molecular Biosciences, University of California, Davis, Davis, California, United States of America
| | - Susan Perlman
- Department of Neurology, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Cecilia Giulivi
- Department of Molecular Biosciences, University of California, Davis, Davis, California, United States of America
| | - Gino A. Cortopassi
- Department of Molecular Biosciences, University of California, Davis, Davis, California, United States of America
| |
Collapse
|
34
|
Muralidharan M, Mitra A, Maity D, Pal D, Mandal AK. Structural analysis of glutathionyl hemoglobin using native mass spectrometry. J Struct Biol 2019; 208:107386. [PMID: 31518635 DOI: 10.1016/j.jsb.2019.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 09/05/2019] [Accepted: 09/07/2019] [Indexed: 01/18/2023]
Abstract
Glutathionylation is an example of reversible post-translation modification of proteins where free and accessible cysteine residues of proteins undergo thiol-disulfide exchange with oxidized glutathione (GSSG). In general, glutathionylation occurs under the condition of elevated oxidative stress in vivo. In human hemoglobin, Cys93 residue of β globin chain was found to undergo this oxidative modification. Glutathionyl hemoglobin (GSHb) was reported to act as a biomarker of oxidative stress under several clinical conditions such as chronic renal failure, iron deficiency anemia, hyperlipidemia, diabetes mellitus, Friedreich's ataxia, atherosclerosis. Previously we showed that the functional abnormality associated with six-fold tighter oxygen binding of GSHb supposedly attributed to the conformational transition of the deoxy state of GSHb towards oxy hemoglobin like conformation. In the present study, we investigated the structural integrity and overall architecture of the quaternary structure of GSHb using native mass spectrometry and ion mobility mass spectrometry platforms. The dissociation equilibrium constants of both tetramer/dimer (Kd1) and dimer/monomer equilibrium (Kd2) was observed to increase by 1.91 folds and 3.64 folds respectively. However, the collision cross-section area of the tetrameric hemoglobin molecule remained unchanged upon glutathionylation. The molecular dynamics simulation data of normal human hemoglobin and GSHb was employed to support our experimental findings.
Collapse
Affiliation(s)
- Monita Muralidharan
- Clinical Proteomics Unit, Division of Molecular Medicine, St. John's Research Institute, 100 ft Road, Koramangala, Bangalore 560034, India
| | - Amrita Mitra
- Clinical Proteomics Unit, Division of Molecular Medicine, St. John's Research Institute, 100 ft Road, Koramangala, Bangalore 560034, India
| | - Dibyajyoti Maity
- IISc Mathematics Initiative, Indian Institute of Science, Bangalore 560012, India
| | - Debnath Pal
- Bioinformatics Centre, Department of Computational & Data Sciences, Indian Institute of Science, Bangalore 560012, India
| | - Amit Kumar Mandal
- Clinical Proteomics Unit, Division of Molecular Medicine, St. John's Research Institute, 100 ft Road, Koramangala, Bangalore 560034, India.
| |
Collapse
|
35
|
Han THL, Camadro JM, Barbault F, Santos R, El Hage Chahine JM, Ha-Duong NT. In Vitro interaction between yeast frataxin and superoxide dismutases: Influence of mitochondrial metals. Biochim Biophys Acta Gen Subj 2019; 1863:883-892. [PMID: 30797804 DOI: 10.1016/j.bbagen.2019.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Friedreich's ataxia results from a decreased expression of the nuclear gene encoding the mitochondrial protein, frataxin. Frataxin participates in the biosynthesis of iron-sulfur clusters and heme cofactors, as well as in iron storage and protection against oxidative stress. How frataxin interacts with the antioxidant defence components is poorly understood. METHODS Therefore, we have investigated by kinetic, thermodynamic and modelling approaches the molecular interactions between yeast frataxin (Yfh1) and superoxide dismutases, Sod1 and Sod2, and the influence of Yfh1 on their enzymatic activities. RESULTS Yfh1 interacts with cytosolic Sod1 with a dissociation constant, Kd = 1.3 ± 0.3 μM, in two kinetic steps. The first step occurs in the 200 ms range and corresponds to the Yfh1-Sod1 interaction, whereas the second is slow and is assumed to be a change in the conformation of the protein-protein adduct. Furthermore, computational investigations confirm the stability of the Yfh1-Sod1 complex. Yfh1 forms two protein complexes with mitochondrial Sod2 with 1:1 and 2:1 Yfh1/Sod2 stoichiometry (Kd1 = 1.05 ± 0.05 and Kd2 = 6.6 ± 0.1 μM). Furthermore, Yfh1 increases the enzymatic activity of Sod1 while slightly affecting that of Sod2. Finally, the stabilities of the protein-protein adducts and the effect of Yfh1 on superoxide dismutase activities depend on the nature of the mitochondrial metal. CONCLUSIONS This work confirms the participation of Yfh1 in cellular defence against oxidative stress.
Collapse
Affiliation(s)
- Thi Hong Lien Han
- Interfaces, Traitements, Organisation et Dynamique des Systèmes (ITODYS), CNRS UMR 7086, Univ Paris Diderot, Sorbonne Paris Cité, 15 rue Jean-Antoine de Baïf, F-75205 Paris Cedex 13, France
| | - Jean-Michel Camadro
- Mitochondries, Métaux et Stress Oxydant, Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, 15 rue Hélène Brion, F-75205 Paris Cedex 13, France
| | - Florent Barbault
- Interfaces, Traitements, Organisation et Dynamique des Systèmes (ITODYS), CNRS UMR 7086, Univ Paris Diderot, Sorbonne Paris Cité, 15 rue Jean-Antoine de Baïf, F-75205 Paris Cedex 13, France
| | - Renata Santos
- Mitochondries, Métaux et Stress Oxydant, Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, 15 rue Hélène Brion, F-75205 Paris Cedex 13, France
| | - Jean-Michel El Hage Chahine
- Interfaces, Traitements, Organisation et Dynamique des Systèmes (ITODYS), CNRS UMR 7086, Univ Paris Diderot, Sorbonne Paris Cité, 15 rue Jean-Antoine de Baïf, F-75205 Paris Cedex 13, France
| | - Nguyet-Thanh Ha-Duong
- Interfaces, Traitements, Organisation et Dynamique des Systèmes (ITODYS), CNRS UMR 7086, Univ Paris Diderot, Sorbonne Paris Cité, 15 rue Jean-Antoine de Baïf, F-75205 Paris Cedex 13, France.
| |
Collapse
|
36
|
Weiland A, Wang Y, Wu W, Lan X, Han X, Li Q, Wang J. Ferroptosis and Its Role in Diverse Brain Diseases. Mol Neurobiol 2018; 56:4880-4893. [PMID: 30406908 DOI: 10.1007/s12035-018-1403-3] [Citation(s) in RCA: 344] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/18/2018] [Indexed: 02/07/2023]
Abstract
Ferroptosis is a recently identified, iron-regulated, non-apoptotic form of cell death. It is characterized by cellular accumulation of lipid reactive oxygen species that ultimately leads to oxidative stress and cell death. Although first identified in cancer cells, ferroptosis has been shown to have significant implications in several neurologic diseases, such as ischemic and hemorrhagic stroke, Alzheimer's disease, and Parkinson's disease. This review summarizes current research on ferroptosis, its underlying mechanisms, and its role in the progression of different neurologic diseases. Understanding the role of ferroptosis could provide valuable information regarding treatment and prevention of these devastating diseases.
Collapse
Affiliation(s)
- Abigail Weiland
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yamei Wang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Neural Regeneration and Repair, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Weihua Wu
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Neural Regeneration and Repair, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xi Lan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Qian Li
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Neural Regeneration and Repair, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Advanced Innovation Center for Human Brain Protection, Captical Medical University, Beijing, 100069, China.
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
37
|
Srivastava D, Kukkuta Sarma GR, Dsouza DS, Muralidharan M, Srinivasan K, Mandal AK. Characterization of residue-specific glutathionylation of CSF proteins in multiple sclerosis - A MS-based approach. Anal Biochem 2018; 564-565:108-115. [PMID: 30367882 DOI: 10.1016/j.ab.2018.10.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/12/2018] [Accepted: 10/14/2018] [Indexed: 12/27/2022]
Abstract
Reduction of a disulfide linkage between cysteine residues in proteins, a standard step in the preanalytical preparation of samples in conventional proteomics approach, presents a challenge to characterize S-glutathionylation of proteins. S-glutathionylation of proteins has been reported in medical conditions associated with high oxidative stress. In the present study, we attempted to characterize glutathionylation of CSF proteins in patients with multiple sclerosis which is associated with high oxidative stress. Using the nano-LC/ESI-MS platform, we adopted a modified proteomics approach and a targeted database search to investigate glutathionylation at the residue level of CSF proteins. Compared to patients with Intracranial hypertension, the following CSF proteins: Extracellular Superoxide dismutase (ECSOD) at Cys195, α1-antitrypsin (A1AT) at Cys232, Phospholipid transfer protein (PLTP) at Cys318, Alpha-2-HS-glycoprotein at Cys340, Ectonucleotide pyrophosphate (ENPP-2) at Cys773, Gelsolin at Cys304, Interleukin-18 (IL-18) at Cys38 and Ig heavy chain V III region POM at Cys22 were found to be glutathionylated in patients with multiple sclerosis during a relapse. ECSOD, A1AT, and PLTP were observed to be glutathionylated at the functionally important cysteine residues. In conclusion, in the present study using a modified proteomics approach we have identified and characterized glutathionylation of CSF proteins in patients with multiple sclerosis.
Collapse
Affiliation(s)
- Deepsikha Srivastava
- Division of Molecular Medicine, Clinical Proteomics Unit, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, 560034, India
| | - Gosala Raja Kukkuta Sarma
- Department of Neurology, St. John's Hospital, St. John's National Academy of Health Sciences, Bangalore, 560034, India
| | - Delon Snehal Dsouza
- Department of Neurology, St. John's Hospital, St. John's National Academy of Health Sciences, Bangalore, 560034, India
| | - Monita Muralidharan
- Division of Molecular Medicine, Clinical Proteomics Unit, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, 560034, India
| | - Krishnamachari Srinivasan
- Department of Psychiatry, St. John's Medical College and Hospital, St. John's National Academy of Health Sciences, Bangalore, 560034, India
| | - Amit Kumar Mandal
- Division of Molecular Medicine, Clinical Proteomics Unit, St. John's Research Institute, St. John's National Academy of Health Sciences, Bangalore, 560034, India.
| |
Collapse
|
38
|
Teskey G, Abrahem R, Cao R, Gyurjian K, Islamoglu H, Lucero M, Martinez A, Paredes E, Salaiz O, Robinson B, Venketaraman V. Glutathione as a Marker for Human Disease. Adv Clin Chem 2018; 87:141-159. [PMID: 30342710 DOI: 10.1016/bs.acc.2018.07.004] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glutathione (GSH), often referred to as "the master antioxidant," participates not only in antioxidant defense systems, but many metabolic processes, and therefore its role cannot be overstated. GSH deficiency causes cellular risk for oxidative damage and thus as expected, GSH imbalance is observed in a wide range of pathological conditions including tuberculosis (TB), HIV, diabetes, cancer, and aging. Consequently, it is not surprising that GSH has attracted the attention of biological researchers and pharmacologists alike as a possible target for medical intervention. Here, we discuss the role GSH plays amongst these pathological conditions to illuminate how it can be used as a marker for human disease.
Collapse
Affiliation(s)
- Garrett Teskey
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Rachel Abrahem
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Ruoqiong Cao
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States; College of life Sciences, Hebei University, Baoding, China
| | - Karo Gyurjian
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Hicret Islamoglu
- Department of Biological Sciences, California State Polytechnic University, Pomona, CA, United States
| | - Mariana Lucero
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Andrew Martinez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Erik Paredes
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Oscar Salaiz
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Brittanie Robinson
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Vishwanath Venketaraman
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States; Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
39
|
Monnier V, Llorens JV, Navarro JA. Impact of Drosophila Models in the Study and Treatment of Friedreich's Ataxia. Int J Mol Sci 2018; 19:E1989. [PMID: 29986523 PMCID: PMC6073496 DOI: 10.3390/ijms19071989] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/26/2018] [Accepted: 07/03/2018] [Indexed: 02/07/2023] Open
Abstract
Drosophila melanogaster has been for over a century the model of choice of several neurobiologists to decipher the formation and development of the nervous system as well as to mirror the pathophysiological conditions of many human neurodegenerative diseases. The rare disease Friedreich’s ataxia (FRDA) is not an exception. Since the isolation of the responsible gene more than two decades ago, the analysis of the fly orthologue has proven to be an excellent avenue to understand the development and progression of the disease, to unravel pivotal mechanisms underpinning the pathology and to identify genes and molecules that might well be either disease biomarkers or promising targets for therapeutic interventions. In this review, we aim to summarize the collection of findings provided by the Drosophila models but also to go one step beyond and propose the implications of these discoveries for the study and cure of this disorder. We will present the physiological, cellular and molecular phenotypes described in the fly, highlighting those that have given insight into the pathology and we will show how the ability of Drosophila to perform genetic and pharmacological screens has provided valuable information that is not easily within reach of other cellular or mammalian models.
Collapse
Affiliation(s)
- Véronique Monnier
- Unité de Biologie Fonctionnelle et Adaptative (BFA), Sorbonne Paris Cité, Université Paris Diderot, UMR8251 CNRS, 75013 Paris, France.
| | - Jose Vicente Llorens
- Department of Genetics, University of Valencia, Campus of Burjassot, 96100 Valencia, Spain.
| | - Juan Antonio Navarro
- Lehrstuhl für Entwicklungsbiologie, Universität Regensburg, 93040 Regensburg, Germany.
| |
Collapse
|
40
|
Nwidu LL, Oboma YI, Elmorsy E, Carter WG. Hepatoprotective effect of hydromethanolic leaf extract of Musanga cecropioides (Urticaceae) on carbon tetrachloride-induced liver injury and oxidative stress. J Taibah Univ Med Sci 2018; 13:344-354. [PMID: 31435346 PMCID: PMC6694957 DOI: 10.1016/j.jtumed.2018.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/05/2018] [Accepted: 04/12/2018] [Indexed: 01/31/2023] Open
Abstract
Objective Natural antioxidant products are gaining popularity as treatments for various pathological liver injuries. Musanga cecropioides (Urticaceae) leaf extract is used in ethnomedicine for the management of jaundice and other hepatic ailments in Ibibio, Nigeria. This study evaluated the hepatoprotective and antioxidant effects of M. cecropioides hydromethanolic leaf (MCHL) extract against carbon tetrachloride (CCl4)-induced hepatotoxicity in rats. Methods Liver damage was induced by administering CCl4 dissolved in liquid paraffin (2 mL/kg bw 1:1 intraperitoneally) after pretreatment with MCHL extract for 7 days. Thereafter, acute hepatotoxicity was evaluated in 36 Wistar rats divided into six groups (A–F) of six animals each. Group A served as the negative control; B received CCl4 1 mL/kg only; C–E received 70.7, 141.4, and 282.8 mg/kg MCHL extract, respectively; and F received silymarin 100 mg/kg daily for 7 days by oral gavage. After 48 h, the rats were sacrificed, and samples obtained from them were assayed for histological and biochemical biomarkers of hepatotoxicity. Results The MCHL extracts significantly (p < 0.001–0.05) reduced the increase in aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), conjugated bilirubin (CBIL), and total bilirubin (TBIL) levels induced by CCl4 intoxication. There was no significant alteration in haematological indices or weight following administration of the MCHL extracts. Histopathological examinations revealed mitotic bodies in the 141.4 mg/kg MCHL extract-treated rats, an indication of tissue repair processes. Conclusion The MCHL extract has a dose-specific hepatoprotective effect; hence, the utilisation of this extract for the management of hepatitis requires caution.
Collapse
Affiliation(s)
- Lucky L Nwidu
- Department of Experimental Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, University of Port Harcourt, Choba, Nigeria
| | - Yibala I Oboma
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Egypt
| | - Ekramy Elmorsy
- Department of Medical Laboratory Science, Faculty of Basic Medical Sciences, College Health Sciences, Niger Delta University, Wilberforce Island, Bayelsa State, Nigeria
| | - Wayne G Carter
- School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, UK
| |
Collapse
|
41
|
Nwidu LL, Elmorsy E, Oboma YI, Carter WG. Hepatoprotective and antioxidant activities of Spondias mombin leaf and stem extracts against carbon tetrachloride-induced hepatotoxicity. J Taibah Univ Med Sci 2018; 13:262-271. [PMID: 31435333 PMCID: PMC6695064 DOI: 10.1016/j.jtumed.2018.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/11/2018] [Accepted: 03/14/2018] [Indexed: 12/19/2022] Open
Abstract
Objective Spondias mombin L. is a tree used in folk medicine in Nigeria for the treatment of hepatitis. This study was carried out to comparatively evaluate the hepatoprotective and antioxidant effects of S. mombin leaf and stem (SML and SMS) methanolic extracts in a rat model of carbon tetrachloride (CCl4)-induced hepatotoxicity. Methods Forty-two rats were distributed into seven groups. Groups A and B received water; groups C and D received 500 and 1000 mg/kg SML extract, respectively; groups E and F received 500 and 1000 mg/kg SMS extract, respectively; and group G received 100 mg/kg silymarin. Water, the two extracts, and silymarin were administered daily by oral gavage for 7 days. Hepatotoxicity was induced in groups B to G by the administration of CCl4 once on the seventh day. After 48 h, rats were sacrificed, and tissues and serum samples were examined for histological and biochemical indices of hepatotoxicity. Results Administration of CCl4 resulted in liver injury with significant elevation in the hepatocellular injury markers alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), total bilirubin (TBIL), and conjugated bilirubin (CBIL), associated with a significant reduction in total circulatory protein. Pretreatment with SML and SMS extracts at both doses significantly ameliorated liver injury; lowered ALT, AST, ALP, TBIL, and CBIL levels; elevated cellular glutathione levels as well as catalase and superoxide dismutase activities; and decreased the levels of thiobarbituric acid reactive substances. Conclusion This study provides preliminary evidence supporting the potential therapeutic benefit of S. mombin in xenobiotic-induced hepatotoxicity.
Collapse
Affiliation(s)
- Lucky L Nwidu
- Department of Experimental Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, University of Port Harcourt, Choba, East West Road, Rivers State, Nigeria
| | - Ekramy Elmorsy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, Egypt
| | - Yibala I Oboma
- Department of Medical Laboratory Science, Faculty of Basic Medical Sciences, College of Health Sciences, Wilberforce Island, Bayelsa State, Nigeria
| | - Wayne G Carter
- School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, UK
| |
Collapse
|
42
|
Chandran V, Gao K, Swarup V, Versano R, Dong H, Jordan MC, Geschwind DH. Inducible and reversible phenotypes in a novel mouse model of Friedreich's Ataxia. eLife 2017; 6:e30054. [PMID: 29257745 PMCID: PMC5736353 DOI: 10.7554/elife.30054] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/20/2017] [Indexed: 12/13/2022] Open
Abstract
Friedreich's ataxia (FRDA), the most common inherited ataxia, is caused by recessive mutations that reduce the levels of frataxin (FXN), a mitochondrial iron binding protein. We developed an inducible mouse model of Fxn deficiency that enabled us to control the onset and progression of disease phenotypes by the modulation of Fxn levels. Systemic knockdown of Fxn in adult mice led to multiple phenotypes paralleling those observed in human patients across multiple organ systems. By reversing knockdown after clinical features appear, we were able to determine to what extent observed phenotypes represent reversible cellular dysfunction. Remarkably, upon restoration of near wild-type FXN levels, we observed significant recovery of function, associated pathology and transcriptomic dysregulation even after substantial motor dysfunction and pathology were observed. This model will be of broad utility in therapeutic development and in refining our understanding of the relative contribution of reversible cellular dysfunction at different stages in disease.
Collapse
Affiliation(s)
- Vijayendran Chandran
- Program in Neurogenetics, Department of Neurology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Kun Gao
- Program in Neurogenetics, Department of Neurology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Vivek Swarup
- Program in Neurogenetics, Department of Neurology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Revital Versano
- Program in Neurogenetics, Department of Neurology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Hongmei Dong
- Program in Neurogenetics, Department of Neurology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Maria C Jordan
- Department of Physiology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
- Department of Human Genetics, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesUnited States
| |
Collapse
|
43
|
Enns GM, Cohen BH. Clinical Trials in Mitochondrial Disease. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2017. [DOI: 10.1177/2326409817733013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Gregory M. Enns
- Department of Pediatrics, Division of Medical Genetics, Stanford University, Stanford, CA, USA
| | - Bruce H. Cohen
- Department of Pediatrics, NeuroDevelopmental Science Center and Mitochondrial Research Center, Akron Children's Hospital, Akron, OH
| |
Collapse
|
44
|
Coles LD, Tuite PJ, Öz G, Mishra UR, Kartha RV, Sullivan KM, Cloyd JC, Terpstra M. Repeated-Dose Oral N-Acetylcysteine in Parkinson's Disease: Pharmacokinetics and Effect on Brain Glutathione and Oxidative Stress. J Clin Pharmacol 2017; 58:158-167. [PMID: 28940353 DOI: 10.1002/jcph.1008] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/01/2017] [Indexed: 12/19/2022]
Abstract
Parkinson's disease (PD) is associated with oxidative stress and decreased nigral glutathione (GSH), suggesting that therapies that boost GSH may have a disease-modifying effect. Intravenous administration of a high dose of N-acetylcysteine (NAC), a well-known antioxidant and GSH precursor, increases blood and brain GSH in individuals with PD and with Gaucher disease and in healthy controls. To characterize the pharmacokinetics of repeated high oral doses of NAC and their effect on brain and blood oxidative stress measures, we conducted a 4-week open-label prospective study of oral NAC in individuals with PD (n = 5) and in healthy controls (n = 3). Brain GSH was measured in the occipital cortex using 1 H-MRS at 3 and 7 tesla before and after 28 days of 6000 mg NAC/day. Blood was collected prior to dosing and at predetermined collection times before and after the last dose to assess NAC, cysteine, GSH, catalase, malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE) concentrations and the reduced-to-oxidized GSH ratio (GSH/ glutathione disulfide [GSSG]). Symptomatic adverse events were reported by 3 of the 5 subjects with PD. NAC plasma concentration-time profiles were described by a first-order absorption, 1-compartment pharmacokinetic model. Although peripheral antioxidant measures (catalase and GSH/GSSG) increased significantly relative to baseline, indicators of oxidative damage, that is, measures of lipid peroxidation (4-HNE and MDA) were unchanged. There were no significant increases in brain GSH, which may be related to low oral NAC bioavailability and small fractional GSH/GSSG blood responses. Additional studies are needed to further characterize side effects and explore the differential effects of NAC on measures of antioxidant defense and oxidative damage.
Collapse
Affiliation(s)
- Lisa D Coles
- Center for Orphan Drug Research, Department of Experimental & Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Paul J Tuite
- Department of Neurology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Gülin Öz
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Usha R Mishra
- Center for Orphan Drug Research, Department of Experimental & Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Reena V Kartha
- Center for Orphan Drug Research, Department of Experimental & Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Kathleen M Sullivan
- Center for Orphan Drug Research, Department of Experimental & Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - James C Cloyd
- Center for Orphan Drug Research, Department of Experimental & Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Melissa Terpstra
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, Medical School, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
45
|
Enns GM, Cowan TM. Glutathione as a Redox Biomarker in Mitochondrial Disease-Implications for Therapy. J Clin Med 2017; 6:jcm6050050. [PMID: 28467362 PMCID: PMC5447941 DOI: 10.3390/jcm6050050] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/24/2017] [Accepted: 04/27/2017] [Indexed: 01/01/2023] Open
Abstract
Technical advances in the ability to measure mitochondrial dysfunction are providing new insights into mitochondrial disease pathogenesis, along with new tools to objectively evaluate the clinical status of mitochondrial disease patients. Glutathione (l-ϒ-glutamyl-l-cysteinylglycine) is the most abundant intracellular thiol, and the intracellular redox state, as reflected by levels of oxidized (GSSG) and reduced (GSH) glutathione, as well as the GSH/GSSG ratio, is considered to be an important indication of cellular health. The ability to quantify mitochondrial dysfunction in an affected patient will not only help with routine care, but also improve rational clinical trial design aimed at developing new therapies. Indeed, because multiple disorders have been associated with either primary or secondary deficiency of the mitochondrial electron transport chain and redox imbalance, developing mitochondrial therapies that have the potential to improve the intracellular glutathione status has been a focus of several clinical trials over the past few years. This review will also discuss potential therapies to increase intracellular glutathione with a focus on EPI-743 (α-tocotrienol quinone), a compound that appears to have the ability to modulate the activity of oxidoreductases, in particular NAD(P)H:quinone oxidoreductase 1.
Collapse
Affiliation(s)
- Gregory M Enns
- Departments of Pediatrics and Pathology, Stanford University, 300 Pasteur Drive, H-315, Stanford, CA 94005-5208, USA.
| | - Tina M Cowan
- Departments of Pediatrics and Pathology, Stanford University, 300 Pasteur Drive, H-315, Stanford, CA 94005-5208, USA.
| |
Collapse
|
46
|
Calatrava-Ferreras L, Gonzalo-Gobernado R, Reimers D, Herranz AS, Casarejos MJ, Jiménez-Escrig A, Regadera J, Velasco-Martín J, Vallejo-Muñoz M, Díaz-Gil JJ, Bazán E. Liver Growth Factor (LGF) Upregulates Frataxin Protein Expression and Reduces Oxidative Stress in Friedreich's Ataxia Transgenic Mice. Int J Mol Sci 2016; 17:E2066. [PMID: 27941692 PMCID: PMC5187866 DOI: 10.3390/ijms17122066] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/28/2016] [Accepted: 12/06/2016] [Indexed: 01/03/2023] Open
Abstract
Friedreich's ataxia (FA) is a severe disorder with autosomal recessive inheritance that is caused by the abnormal expansion of GAA repeat in intron 1 of FRDA gen. This alteration leads to a partial silencing of frataxin transcription, causing a multisystem disorder disease that includes neurological and non-neurological damage. Recent studies have proven the effectiveness of neurotrophic factors in a number of neurodegenerative diseases. Therefore, we intend to determine if liver growth factor (LGF), which has a demonstrated antioxidant and neuroprotective capability, could be a useful therapy for FA. To investigate the potential therapeutic activity of LGF we used transgenic mice of the FXNtm1MknTg (FXN)YG8Pook strain. In these mice, intraperitoneal administration of LGF (1.6 μg/mouse) exerted a neuroprotective effect on neurons of the lumbar spinal cord and improved cardiac hypertrophy. Both events could be the consequence of the increment in frataxin expression induced by LGF in spinal cord (1.34-fold) and heart (1.2-fold). LGF also upregulated by 2.6-fold mitochondrial chain complex IV expression in spinal cord, while in skeletal muscle it reduced the relation oxidized glutathione/reduced glutathione. Since LGF partially restores motor coordination, we propose LGF as a novel factor that may be useful in the treatment of FA.
Collapse
Affiliation(s)
- Lucía Calatrava-Ferreras
- Service of Neurobiology, Ramón y Cajal Institute for Health Research (IRYCIS), 28034 Madrid, Spain.
| | - Rafael Gonzalo-Gobernado
- Service of Neurobiology, Ramón y Cajal Institute for Health Research (IRYCIS), 28034 Madrid, Spain.
| | - Diana Reimers
- Service of Neurobiology, Ramón y Cajal Institute for Health Research (IRYCIS), 28034 Madrid, Spain.
| | - Antonio S Herranz
- Service of Neurobiology, Ramón y Cajal Institute for Health Research (IRYCIS), 28034 Madrid, Spain.
| | - María J Casarejos
- Service of Neurobiology, Ramón y Cajal Institute for Health Research (IRYCIS), 28034 Madrid, Spain.
| | | | - Javier Regadera
- Departamento de Anatomía, Histología y Neurociencia Facultad de Medicina Universidad Autónoma de Madrid, 28400 Madrid, Spain.
| | - Juan Velasco-Martín
- Departamento de Anatomía, Histología y Neurociencia Facultad de Medicina Universidad Autónoma de Madrid, 28400 Madrid, Spain.
| | - Manuela Vallejo-Muñoz
- Service of Neurobiology, Ramón y Cajal Institute for Health Research (IRYCIS), 28034 Madrid, Spain.
| | - Juan José Díaz-Gil
- Service of Neurobiology, Ramón y Cajal Institute for Health Research (IRYCIS), 28034 Madrid, Spain.
| | - Eulalia Bazán
- Service of Neurobiology, Ramón y Cajal Institute for Health Research (IRYCIS), 28034 Madrid, Spain.
| |
Collapse
|
47
|
Tamarit J, Obis È, Ros J. Oxidative stress and altered lipid metabolism in Friedreich ataxia. Free Radic Biol Med 2016; 100:138-146. [PMID: 27296838 DOI: 10.1016/j.freeradbiomed.2016.06.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 12/31/2022]
Abstract
Friedreich ataxia is a genetic disease caused by the deficiency of frataxin, a mitochondrial protein. Frataxin deficiency impacts in the cell physiology at several levels. One of them is oxidative stress with consequences in terms of protein dysfunctions and metabolic alterations. Among others, alterations in lipid metabolism have been observed in several models of the disease. In this review we summarize the current knowledge of the molecular basis of the disease, the relevance of oxidative stress and the therapeutic strategies based on reduction of mitochondrial reactive oxygen species production. Finally, we will focus the interest in alterations of lipid metabolism as a consequence of mitochondrial dysfunction and describe the therapeutic approaches based on targeting lipid metabolism.
Collapse
Affiliation(s)
- Jordi Tamarit
- Departament de Ciències Mèdiques Bàsiques, IRB-Lleida, Universitat de Lleida, Lleida, Spain
| | - Èlia Obis
- Departament de Ciències Mèdiques Bàsiques, IRB-Lleida, Universitat de Lleida, Lleida, Spain
| | - Joaquim Ros
- Departament de Ciències Mèdiques Bàsiques, IRB-Lleida, Universitat de Lleida, Lleida, Spain.
| |
Collapse
|
48
|
Hayashi G, Cortopassi G. Lymphoblast Oxidative Stress Genes as Potential Biomarkers of Disease Severity and Drug Effect in Friedreich's Ataxia. PLoS One 2016; 11:e0153574. [PMID: 27078885 PMCID: PMC4831832 DOI: 10.1371/journal.pone.0153574] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 03/31/2016] [Indexed: 11/25/2022] Open
Abstract
There is no current approved therapy for the ultimately lethal neuro- and cardio-degenerative disease Friedreich's ataxia (FA). Finding minimally-invasive molecular biomarkers of disease progression and drug effect could support smaller, shorter clinical trials. Since we and others have noted a deficient oxidative stress response in FA, we investigated the expression of 84 genes involved in oxidative stress, signaling, and protection in control and FA lymphoblasts ranging from 460 to 1122 GAA repeats. Several antioxidant genes responded in a dose-dependent manner to frataxin expression at the mRNA and protein levels, which is inversely correlated with disease progression and severity. We tested the effect of experimental Friedreich's ataxia therapies dimethyl fumarate (DMF) and type 1 histone deacetylase inhibitor (HDACi) on biomarker mRNA expression. We observed that exposure of lymphoblasts to DMF and HDACi dose-dependently unsilenced frataxin expression and restored the potential biomarkers NCF2 and PDLIM1 expression to control levels. We suggest that in addition to frataxin expression, blood lymphoblast levels of NCF2 and PDLIM1 could be useful biomarkers for disease progression and drug effect in future clinical trials of Friedreich's ataxia.
Collapse
Affiliation(s)
- Genki Hayashi
- Department of Molecular Biosciences, University of California Davis, Davis, California, United States of America
| | - Gino Cortopassi
- Department of Molecular Biosciences, University of California Davis, Davis, California, United States of America
| |
Collapse
|
49
|
Hayashi G, Cortopassi G. Oxidative stress in inherited mitochondrial diseases. Free Radic Biol Med 2015; 88:10-7. [PMID: 26073122 PMCID: PMC4593728 DOI: 10.1016/j.freeradbiomed.2015.05.039] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/10/2015] [Accepted: 05/26/2015] [Indexed: 12/22/2022]
Abstract
Mitochondria are a source of reactive oxygen species (ROS). Mitochondrial diseases are the result of inherited defects in mitochondrially expressed genes. One potential pathomechanism for mitochondrial disease is oxidative stress. Oxidative stress can occur as the result of increased ROS production or decreased ROS protection. The role of oxidative stress in the five most common inherited mitochondrial diseases, Friedreich ataxia, LHON, MELAS, MERRF, and Leigh syndrome (LS), is discussed. Published reports of oxidative stress involvement in the pathomechanisms of these five mitochondrial diseases are reviewed. The strongest evidence for an oxidative stress pathomechanism among the five diseases was for Friedreich ataxia. In addition, a meta-analysis was carried out to provide an unbiased evaluation of the role of oxidative stress in the five diseases, by searching for "oxidative stress" citation count frequency for each disease. Of the five most common mitochondrial diseases, the strongest support for oxidative stress is for Friedreich ataxia (6.42%), followed by LHON (2.45%), MELAS (2.18%), MERRF (1.71%), and LS (1.03%). The increased frequency of oxidative stress citations was significant relative to the mean of the total pool of five diseases (p<0.01) and the mean of the four non-Friedreich diseases (p<0.0001). Thus there is support for oxidative stress in all five most common mitochondrial diseases, but the strongest, significant support is for Friedreich ataxia.
Collapse
Affiliation(s)
- Genki Hayashi
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA
| | - Gino Cortopassi
- Department of Molecular Biosciences, University of California, Davis, CA 95616, USA.
| |
Collapse
|
50
|
Ehrhart J, Smith AJ, Kuzmin-Nichols N, Zesiewicz TA, Jahan I, Shytle RD, Kim SH, Sanberg CD, Vu TH, Gooch CL, Sanberg PR, Garbuzova-Davis S. Humoral factors in ALS patients during disease progression. J Neuroinflammation 2015; 12:127. [PMID: 26126965 PMCID: PMC4487852 DOI: 10.1186/s12974-015-0350-4] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 06/19/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting upper and lower motor neurons in the CNS and leading to paralysis and death. There are currently no effective treatments for ALS due to the complexity and heterogeneity of factors involved in motor neuron degeneration. A complex of interrelated effectors have been identified in ALS, yet systemic factors indicating and/or reflecting pathological disease developments are uncertain. The purpose of the study was to identify humoral effectors as potential biomarkers during disease progression. METHODS Thirteen clinically definite ALS patients and seven non-neurological controls enrolled in the study. Peripheral blood samples were obtained from each ALS patient and control at two visits separated by 6 months. The Revised ALS Functional Rating Scale (ALSFRS-R) was used to evaluate overall ALS-patient functional status at each visit. Eleven humoral factors were analyzed in sera. Cytokine levels (GM-CSF, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, and TNF-α) were determined using the Bio-Rad Bio-Plex® Luminex 200 multiplex assay system. Nitrite, a breakdown product of NO, was quantified using a Griess Reagent System. Glutathione (GSH) concentrations were measured using a Glutathione Fluorometric Assay Kit. RESULTS ALS patients had ALSFRS-R scores of 30.5 ± 1.9 on their first visit and 27.3 ± 2.7 on the second visit, indicating slight disease progression. Serum multiplex cytokine panels revealed statistically significant changes in IL-2, IL-5, IL-6, and IL-8 levels in ALS patients depending on disease status at each visit. Nitrite serum levels trended upwards in ALS patients while serum GSH concentrations were drastically decreased in sera from ALS patients versus controls at both visits. CONCLUSIONS Our results demonstrated a systemic pro-inflammatory state and impaired antioxidant system in ALS patients during disease progression. Increased levels of pro-inflammatory IL-6, IL-8, and nitrite and significantly decreased endogenous antioxidant GSH levels could identify these humoral constituents as systemic biomarkers for ALS. However, systemic changes in IL-2, IL-5, and IL-6 levels determined between visits in ALS patients might indicate adaptive immune system responses dependent on current disease stage. These novel findings, showing dynamic changes in humoral effectors during disease progression, could be important for development of an effective treatment for ALS.
Collapse
Affiliation(s)
| | - Adam J Smith
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | | | - Theresa A Zesiewicz
- Department of Neurology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | - Israt Jahan
- Department of Neurology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | - R Douglas Shytle
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA. .,Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL, 33612, USA.
| | - Seol-Hee Kim
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | | | - Tuan H Vu
- Department of Neurology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | - Clifton L Gooch
- Department of Neurology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | - Paul R Sanberg
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA. .,Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL, 33612, USA. .,Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA. .,Department of Psychiatry, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| | - Svitlana Garbuzova-Davis
- Center of Excellence for Aging & Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA. .,Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL, 33612, USA. .,Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA. .,Department of Pathology and Cell Biology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|